ϟ

W.H. Wilson Tang

Here are all the papers by W.H. Wilson Tang that you can download and read on OA.mg.
W.H. Wilson Tang’s last known institution is . Download W.H. Wilson Tang PDFs here.

Claim this Profile →
DOI: 10.1016/j.jacc.2013.05.019
2013
Cited 6,986 times
2013 ACCF/AHA Guideline for the Management of Heart Failure
Jeffrey L. Anderson, MD, FACC, FAHA, Chair; Alice K. Jacobs, MD, FACC, FAHA, Immediate Past Chair[‡‡][1]; Jonathan L. Halperin, MD, FACC, FAHA, Chair-Elect; Nancy M. Albert, PhD, CCNS, CCRN, FAHA; Biykem Bozkurt, MD, PhD, FACC, FAHA; Ralph G. Brindis, MD, MPH, MACC; Mark A. Creager, MD, FACC,
DOI: 10.1038/nature09922
2011
Cited 4,291 times
Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease
Metabolomics studies hold promise for the discovery of pathways linked to disease processes. Cardiovascular disease (CVD) represents the leading cause of death and morbidity worldwide. Here we used a metabolomics approach to generate unbiased small-molecule metabolic profiles in plasma that predict risk for CVD. Three metabolites of the dietary lipid phosphatidylcholine—choline, trimethylamine N-oxide (TMAO) and betaine—were identified and then shown to predict risk for CVD in an independent large clinical cohort. Dietary supplementation of mice with choline, TMAO or betaine promoted upregulation of multiple macrophage scavenger receptors linked to atherosclerosis, and supplementation with choline or TMAO promoted atherosclerosis. Studies using germ-free mice confirmed a critical role for dietary choline and gut flora in TMAO production, augmented macrophage cholesterol accumulation and foam cell formation. Suppression of intestinal microflora in atherosclerosis-prone mice inhibited dietary-choline-enhanced atherosclerosis. Genetic variations controlling expression of flavin monooxygenases, an enzymatic source of TMAO, segregated with atherosclerosis in hyperlipidaemic mice. Discovery of a relationship between gut-flora-dependent metabolism of dietary phosphatidylcholine and CVD pathogenesis provides opportunities for the development of new diagnostic tests and therapeutic approaches for atherosclerotic heart disease. Stanley Hazen and colleagues show that gut flora can influence cardiovascular disease by metabolizing a dietary phospholipid. A targeted metabolomics approach was used to identify plasma metabolites whose levels predict future risk for experiencing a non-fatal heart attack, stroke or death in subjects undergoing cardiac evaluation. Plasma levels of three metabolites of dietary phosphatidylcholine — choline, betaine and trimethylamine N-oxide (TMAO) — are associated with increased risk of cardiovascular disease. The gut flora is known to have a role in TMAO formation from choline. In addition, experiments in atherosclerosis-prone mice show that dietary choline enhances macrophage foam-cell formation and lesion formation — but not if the gut flora is depleted with antibiotics. This work suggests new diagnostic and therapeutic approaches for atherosclerotic heart disease. This paper shows that gut flora can influence cardiovascular disease, by metabolizing a dietary phospholipid. Using a metabolomics approach it is found that plasma levels of three metabolites of dietary phosphatidylcholine—choline, betaine and TMAO—are associated with increased risk of cardiovascular disease in humans. The gut flora is known to have a role in TMAO formation from choline, and this paper shows that dietary choline supplementation enhances macrophage foam cell formation and lesion formation in atherosclerosis-prone mice, but not if the gut flora are depleted with antibiotics.
DOI: 10.1038/nm.3145
2013
Cited 3,385 times
Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes atherosclerosis
Intestinal microbiota metabolism of choline and phosphatidylcholine produces trimethylamine (TMA), which is further metabolized to a proatherogenic species, trimethylamine-N-oxide (TMAO). We demonstrate here that metabolism by intestinal microbiota of dietary L-carnitine, a trimethylamine abundant in red meat, also produces TMAO and accelerates atherosclerosis in mice. Omnivorous human subjects produced more TMAO than did vegans or vegetarians following ingestion of L-carnitine through a microbiota-dependent mechanism. The presence of specific bacterial taxa in human feces was associated with both plasma TMAO concentration and dietary status. Plasma L-carnitine levels in subjects undergoing cardiac evaluation (n = 2,595) predicted increased risks for both prevalent cardiovascular disease (CVD) and incident major adverse cardiac events (myocardial infarction, stroke or death), but only among subjects with concurrently high TMAO levels. Chronic dietary L-carnitine supplementation in mice altered cecal microbial composition, markedly enhanced synthesis of TMA and TMAO, and increased atherosclerosis, but this did not occur if intestinal microbiota was concurrently suppressed. In mice with an intact intestinal microbiota, dietary supplementation with TMAO or either carnitine or choline reduced in vivo reverse cholesterol transport. Intestinal microbiota may thus contribute to the well-established link between high levels of red meat consumption and CVD risk.
DOI: 10.1161/cir.0b013e31829e8807
2013
Cited 2,605 times
2013 ACCF/AHA Guideline for the Management of Heart Failure: Executive Summary
HomeCirculationVol. 128, No. 162013 ACCF/AHA Guideline for the Management of Heart Failure: Executive Summary Free AccessResearch ArticlePDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toFree AccessResearch ArticlePDF/EPUB2013 ACCF/AHA Guideline for the Management of Heart Failure: Executive SummaryA Report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines Clyde W. Yancy, MD, MSc, FACC, FAHA, Chair, Mariell Jessup, MD, FACC, FAHA, Vice Chair, Biykem Bozkurt, MD, PhD, FACC, FAHA, Javed Butler, MBBS, FACC, FAHA, Donald E. CaseyJr, MD, MPH, MBA, FACP, FAHA, Mark H. Drazner, MD, MSc, FACC, FAHA, Gregg C. Fonarow, MD, FACC, FAHA, Stephen A. Geraci, MD, FACC, FAHA, FCCP, Tamara Horwich, MD, FACC, James L. Januzzi, MD, FACC, Maryl R. Johnson, MD, FACC, FAHA, Edward K. Kasper, MD, FACC, FAHA, Wayne C. Levy, MD, FACC, Frederick A. Masoudi, MD, MSPH, FACC, FAHA, Patrick E. McBride, MD, MPH, FACC, John J.V. McMurray, MD, FACC, Judith E. Mitchell, MD, FACC, FAHA, Pamela N. Peterson, MD, MSPH, FACC, FAHA, Barbara Riegel, DNSc, RN, FAHA, Flora Sam, MD, FACC, FAHA, Lynne W. Stevenson, MD, FACC, W.H. Wilson Tang, MD, FACC, Emily J. Tsai, MD, FACC and Bruce L. Wilkoff, MD, FACC, FHRS Clyde W. YancyClyde W. Yancy Search for more papers by this author , Mariell JessupMariell Jessup Search for more papers by this author , Biykem BozkurtBiykem Bozkurt Search for more papers by this author , Javed ButlerJaved Butler Search for more papers by this author , Donald E. CaseyJrDonald E. CaseyJr Search for more papers by this author , Mark H. DraznerMark H. Drazner Search for more papers by this author , Gregg C. FonarowGregg C. Fonarow Search for more papers by this author , Stephen A. GeraciStephen A. Geraci Search for more papers by this author , Tamara HorwichTamara Horwich Search for more papers by this author , James L. JanuzziJames L. Januzzi Search for more papers by this author , Maryl R. JohnsonMaryl R. Johnson Search for more papers by this author , Edward K. KasperEdward K. Kasper Search for more papers by this author , Wayne C. LevyWayne C. Levy Search for more papers by this author , Frederick A. MasoudiFrederick A. Masoudi Search for more papers by this author , Patrick E. McBridePatrick E. McBride Search for more papers by this author , John J.V. McMurrayJohn J.V. McMurray Search for more papers by this author , Judith E. MitchellJudith E. Mitchell Search for more papers by this author , Pamela N. PetersonPamela N. Peterson Search for more papers by this author , Barbara RiegelBarbara Riegel Search for more papers by this author , Flora SamFlora Sam Search for more papers by this author , Lynne W. StevensonLynne W. Stevenson Search for more papers by this author , W.H. Wilson TangW.H. Wilson Tang Search for more papers by this author , Emily J. TsaiEmily J. Tsai Search for more papers by this author and Bruce L. WilkoffBruce L. Wilkoff Search for more papers by this author Originally published5 Jun 2013https://doi.org/10.1161/CIR.0b013e31829e8807Circulation. 2013;128:1810–1852Other version(s) of this articleYou are viewing the most recent version of this article. Previous versions: January 1, 2013: Previous Version 1 Table of ContentsPreamble 18111. Introduction 18141.1. Methodology and Evidence Review 18141.2. Organization of the Writing Committee 18141.3. Document Review and Approval 18141.4. Scope of This Guideline With Reference to Other Relevant Guidelines or Statements 18142. Definition of HF 18143. HF Classifications 18164. Epidemiology 18165. Initial and Serial Evaluation of the HF Patient: Recommendations 18175.1. Clinical Evaluation 18175.1.1. History and Physical Examination 18175.1.2. Risk Scoring 18175.2. Diagnostic Tests 18175.3. Biomarkers 18185.4. Noninvasive Cardiac Imaging 18185.5. Invasive Evaluation 18196. Treatment of Stages A to D: Recommendations 18206.1. Stage A 18206.2. Stage B 18206.3. Stage C 18216.3.1. Nonpharmacological Interventions 18216.3.2. Pharmacological Treatment for Stage C HFrEF 18216.3.3. Pharmacological Treatment for Stage C HFpEF 18246.3.4. Device Therapy for Stage C HFrEF 18266.4. Stage D18286.4.1. Water Restriction 18296.4.2. Inotropic Support 18296.4.3. Mechanical Circulatory Support 18306.4.4. Cardiac Transplantation 18307. The Hospitalized Patient: Recommendations 18317.1. Precipitating Causes of Decompensated HF 18317.2. Maintenance of GDMT During Hospitalization 18317.3. Diuretics in Hospitalized Patients 18327.4. Renal Replacement Therapy—Ultrafiltration 18327.5. Parenteral Therapy in Hospitalized HF 18337.6. Venous Thromboembolism Prophylaxis in Hospitalized Patients 18337.7. Arginine Vasopressin Antagonists 18337.8. Inpatient and Transitions of Care 18338. Important Comorbidities in HF 18349. Surgical/Percutaneous/Transcatheter Interventional Treatments of HF: Recommendations 183410. Coordinating Care for Patients With Chronic HF: Recommendations 183511. Quality Metrics/Performance Measures: Recommendations 183512. Evidence Gaps and Future Research Directions1835References 1837Appendix 1. Author Relationships With Industry and Other Entities (Relevant) 1846Appendix 2. Reviewer Relationships With Industry and Other Entities (Relevant) 1849PreambleThe medical profession should play a central role in evaluating the evidence related to drugs, devices, and procedures for the detection, management, and prevention of disease. When properly applied, expert analysis of available data on the benefits and risks of these therapies and procedures can improve the quality of care, optimize patient outcomes, and favorably affect costs by focusing resources on the most effective strategies. An organized and directed approach to a thorough review of evidence has resulted in the production of clinical practice guidelines that assist clinicians in selecting the best management strategy for an individual patient. Moreover, clinical practice guidelines can provide a foundation for other applications, such as performance measures, appropriate use criteria, and both quality improvement and clinical decision support tools.The American College of Cardiology Foundation (ACCF) and the American Heart Association (AHA) have jointly produced guidelines in the area of cardiovascular disease since 1980. The ACCF/AHA Task Force on Practice Guidelines (Task Force), charged with developing, updating, and revising practice guidelines for cardiovascular diseases and procedures, directs and oversees this effort. Writing committees are charged with regularly reviewing and evaluating all available evidence to develop balanced, patient-centric recommendations for clinical practice.Experts in the subject under consideration are selected by the ACCF and AHA to examine subject-specific data and write guidelines in partnership with representatives from other medical organizations and specialty groups. Writing committees are asked to perform a literature review; weigh the strength of evidence for or against particular tests, treatments, or procedures; and include estimates of expected outcomes where such data exist. Patient-specific modifiers, comorbidities, and issues of patient preference that may influence the choice of tests or therapies are considered. When available, information from studies on cost is considered, but data on efficacy and outcomes constitute the primary basis for the recommendations contained herein.In analyzing the data and developing recommendations and supporting text, the writing committee uses evidence-based methodologies developed by the Task Force.1 The Class of Recommendation (COR) is an estimate of the size of the treatment effect considering risks versus benefits in addition to evidence and/or agreement that a given treatment or procedure is or is not useful/effective or in some situations may cause harm. The Level of Evidence (LOE) is an estimate of the certainty or precision of the treatment effect. The writing committee reviews and ranks evidence supporting each recommendation with the weight of evidence ranked as LOE A, B, or C according to specific definitions that are included in Table 1. Studies are identified as observational, retrospective, prospective, or randomized where appropriate. For certain conditions for which inadequate data are available, recommendations are based on expert consensus and clinical experience and are ranked as LOE C. When recommendations at LOE C are supported by historical clinical data, appropriate references (including clinical reviews) are cited if available. For issues for which sparse data are available, a survey of current practice among the clinicians on the writing committee is the basis for LOE C recommendations and no references are cited. The schema for COR and LOE are summarized in Table 1, which also provides suggested phrases for writing recommendations within each COR. A new addition to this methodology is separation of the Class III recommendations to delineate whether the recommendation is determined to be of “no benefit” or is associated with “harm” to the patient. In addition, in view of the increasing number of comparative effectiveness studies, comparator verbs and suggested phrases for writing recommendations for the comparative effectiveness of one treatment or strategy versus another have been added for COR I and IIa, LOE A or B only.Table 1. Applying Classification of Recommendation and Level of EvidenceTable 1. Applying Classification of Recommendation and Level of EvidenceIn view of the advances in medical therapy across the spectrum of cardiovascular diseases, the Task Force has designated the term guideline-directed medical therapy (GDMT) to represent optimal medical therapy as defined by ACCF/AHA guideline−recommended therapies (primarily Class I). This new term, GDMT, will be used herein and throughout all future guidelines.Because the ACCF/AHA practice guidelines address patient populations (and clinicians) residing in North America, drugs that are not currently available in North America are discussed in the text without a specific COR. For studies performed in large numbers of subjects outside North America, each writing committee reviews the potential influence of different practice patterns and patient populations on the treatment effect and relevance to the ACCF/AHA target population to determine whether the findings should inform a specific recommendation.The ACCF/AHA practice guidelines are intended to assist clinicians in clinical decision making by describing a range of generally acceptable approaches to the diagnosis, management, and prevention of specific diseases or conditions. The guidelines attempt to define practices that meet the needs of most patients in most circumstances. The ultimate judgment regarding care of a particular patient must be made by the clinician and patient in light of all the circumstances presented by that patient. As a result, situations may arise for which deviations from these guidelines may be appropriate. Clinical decision making should involve consideration of the quality and availability of expertise in the area where care is provided. When these guidelines are used as the basis for regulatory or payer decisions, the goal should be improvement in quality of care. The Task Force recognizes that situations arise in which additional data are needed to inform patient care more effectively; these areas will be identified within each respective guideline when appropriate.Prescribed courses of treatment in accordance with these recommendations are effective only if followed. Because lack of patient understanding and adherence may adversely affect outcomes, clinicians should make every effort to engage the patient’s active participation in prescribed medical regimens and lifestyles. In addition, patients should be informed of the risks, benefits, and alternatives to a particular treatment and be involved in shared decision making whenever feasible, particularly for COR IIa and IIb, for which the benefit-to-risk ratio may be lower.The Task Force makes every effort to avoid actual, potential, or perceived conflicts of interest that may arise as a result of industry relationships or personal interests among the members of the writing committee. All writing committee members and peer reviewers of the guideline are required to disclose all current healthcare-related relationships, including those existing 12 months before initiation of the writing effort. In December 2009, the ACCF and AHA implemented a new policy for relationship with industry and other entities (RWI) that requires the writing committee chair plus a minimum of 50% of the writing committee to have no relevant RWI (Appendix 1 includes the ACCF/AHA definition of relevance). These statements are reviewed by the Task Force and all members during each conference call and/or meeting of the writing committee and are updated as changes occur. All guideline recommendations require a confidential vote by the writing committee and must be approved by a consensus of the voting members. Members are not permitted to draft or vote on any text or recommendations pertaining to their RWI. Members who recused themselves from voting are indicated in the list of writing committee members, and specific section recusals are noted in Appendix 1. Authors’ and peer reviewers’ RWI pertinent to this guideline are disclosed in Appendixes 1 and 2, respectively. Additionally, to ensure complete transparency, writing committee members’ comprehensive disclosure information—including RWI not pertinent to this document—is available as an online supplement. Comprehensive disclosure information for the Task Force is also available online at http://www.cardiosource.org/en/ACC/About-ACC/Who-We-Are/Leadership/Guidelines-and-Documents-Task-Forces.aspx. The work of writing committees is supported exclusively by the ACCF and AHA without commercial support. Writing committee members volunteered their time for this activity.In an effort to maintain relevance at the point of care for practicing clinicians, the Task Force continues to oversee an ongoing process improvement initiative. As a result, in response to pilot projects, several changes to these guidelines will be apparent, including limited narrative text, a focus on summary and evidence tables (with references linked to abstracts in PubMed), and more liberal use of summary recommendation tables (with references that support LOE) to serve as a quick reference.In April 2011, the Institute of Medicine released 2 reports: Clinical Practice Guidelines We Can Trust and Finding What Works in Health Care: Standards for Systematic Reviews.2,3 It is noteworthy that the ACCF/AHA practice guidelines are cited as being compliant with many of the proposed standards. A thorough review of these reports and of our current methodology is under way, with further enhancements anticipated.The recommendations in this guideline are considered current until they are superseded by a focused update or the full-text guideline is revised. Guidelines are official policy of both the ACCF and AHA. The reader is encouraged to consult the full-text guideline4 for additional guidance and details about heart failure, because the Executive Summary contains only the recommendations.Jeffrey L. Anderson, MD, FACC, FAHAChair, ACCF/AHA Task Force on Practice Guidelines1. Introduction1.1. Methodology and Evidence ReviewThe recommendations listed in this document are, whenever possible, evidence based. An extensive evidence review was conducted through October 2011 and includes selected other references through April 2013. The relevant data are included in evidence tables in the Data Supplement. Searches were extended to studies, reviews, and other evidence conducted in human subjects and that were published in English from PubMed, EMBASE, Cochrane, Agency for Healthcare Research and Quality Reports, and other selected databases relevant to this guideline. Key search words included but were not limited to the following: heart failure, cardiomyopathy, quality of life, mortality, hospitalizations, prevention, biomarkers, hypertension, dyslipidemia, imaging, cardiac catheterization, endomyocardial biopsy, angiotensin-converting enzyme inhibitors, angiotensin-receptor antagonists/blockers, beta blockers, cardiac, cardiac resynchronization therapy, defibrillator, device-based therapy, implantable cardioverter-defibrillator, device implantation, medical therapy, acute decompensated heart failure, preserved ejection fraction, terminal care and transplantation, quality measures, and performance measures. Additionally, the committee reviewed documents related to the subject matter previously published by the ACCF and AHA. References selected and published in this document are representative and not all-inclusive.1.2. Organization of the Writing CommitteeThe committee was composed of physicians and a nurse with broad expertise in the evaluation, care, and management of patients with heart failure (HF). The authors included general cardiologists, HF and transplant specialists, electrophysiologists, general internists, and physicians with methodological expertise. The committee included representatives from the ACCF, AHA, American Academy of Family Physicians, American College of Chest Physicians, American College of Physicians, Heart Rhythm Society, and International Society for Heart and Lung Transplantation.1.3. Document Review and ApprovalThis document was reviewed by 2 official reviewers each nominated by both the ACCF and the AHA, as well as 1 to 2 reviewers each from the American Academy of Family Physicians, American College of Chest Physicians, Heart Rhythm Society, and International Society for Heart and Lung Transplantation, as well as 32 individual content reviewers (including members of the ACCF Adult Congenital and Pediatric Cardiology Council, ACCF Cardiovascular Team Council, ACCF Council on Cardiovascular Care for Older Adults, ACCF Electrophysiology Committee, ACCF Heart Failure and Transplant Council, ACCF Imaging Council, ACCF Prevention Committee, ACCF Surgeons’ Scientific Council, and ACCF Task Force on Appropriate Use Criteria). All information on reviewers’ RWI was distributed to the writing committee and is published in this document (Appendix 2).This document was approved for publication by the governing bodies of the ACCF and AHA and endorsed by the American Association of Cardiovascular and Pulmonary Rehabilitation, American College of Chest Physicians, Heart Rhythm Society, and International Society for Heart and Lung Transplantation.1.4. Scope of This Guideline With Reference to Other Relevant Guidelines or StatementsThis guideline covers multiple management issues for the adult patient with HF. Although there is an abundance of evidence addressing HF, for many important clinical considerations, this writing committee was unable to identify sufficient data to properly inform a recommendation. The writing committee actively worked to reduce the number of LOE “C” recommendations, especially for Class I−recommended therapies. Despite these limitations, it is apparent that much can be done for HF. Adherence to the clinical practice guidelines herein reproduced should lead to improved patient outcomes.Although of increasing importance, children with HF and adults with congenital heart lesions are not specifically addressed in this guideline. The reader is referred to publically available resources to address questions in these areas. However, this guideline does address HF with preserved ejection fraction (EF) in more detail and similarly revisits hospitalized HF. Additional areas of renewed interest are stage D HF, palliative care, transition of care, and quality of care for HF. Certain management strategies appropriate for the patient at risk for HF or already affected by HF are also reviewed in numerous relevant clinical practice guidelines and scientific statements published by the ACCF/AHA Task Force on Practice Guidelines, AHA, ACCF Task Force on Appropriate Use Criteria, European Society of Cardiology, Heart Failure Society of America, and the National Heart, Lung, and Blood Institute. The writing committee saw no need to reiterate the recommendations contained in those guidelines and chose to harmonize recommendations when appropriate and eliminate discrepancies. This is especially the case for device-based therapeutics, where complete alignment between the HF guideline and the device-based therapy guideline was deemed imperative.5 Some recommendations from earlier guidelines have been updated as warranted by new evidence or a better understanding of earlier evidence, whereas others that were no longer accurate or relevant or that were overlapping were modified; recommendations from previous guidelines that were similar or redundant were eliminated or consolidated when possible.The present document recommends a combination of lifestyle modifications and medications that constitute GDMT. GDMT is specifically referenced in the recommendations for treatment of HF (Section 6.3.2). Both for GDMT and other recommended drug treatment regimens, the reader is advised to confirm dosages with product insert material and to evaluate carefully for contraindications and drug-drug interactions. Table 2 is a list of documents deemed pertinent to this effort and is intended for use as a resource; it obviates the need to repeat already extant guideline recommendations. Additional other HF guideline statements are highlighted as well for the purpose of comparison and completeness.Table 2. Associated Guidelines and StatementsTitleOrganizationPublication Year (Reference)Guidelines Guidelines for the Management of Adults With Congenital Heart DiseaseACCF/AHA20086 Guidelines for the Management of Patients With Atrial FibrillationACCF/AHA/HRS20117–9 Guideline for Assessment of Cardiovascular Risk in Asymptomatic AdultsACCF/AHA201010 Guideline for Coronary Artery Bypass Graft SurgeryACCF/AHA201111 Guidelines for Device-Based Therapy of Cardiac Rhythm AbnormalitiesACCF/AHA/HRS20135 Guideline for the Diagnosis and Treatment of Hypertrophic CardiomyopathyACCF/AHA201112 Guideline for Percutaneous Coronary InterventionACCF/AHA/SCAI201113 Secondary Prevention and Risk Reduction Therapy for Patients With Coronary and Other Atherosclerotic Vascular Disease: 2011 UpdateAHA/ACCF201114 Guideline for the Diagnosis and Management of Patients With Stable Ischemic Heart DiseaseACCF/AHA/ACP/AATS/PCNA/SCAI/STS201215 Guideline for the Management of ST-Elevation Myocardial InfarctionACCF/AHA201316 Guidelines for the Management of Patients With Unstable Angina/Non–ST-Elevation Myocardial InfarctionACCF/AHA201317 Guidelines for the Management of Patients With Valvular Heart DiseaseACCF/AHA200818 Comprehensive Heart Failure Practice GuidelineHFSA201019 Guidelines for the Diagnosis and Treatment of Acute and Chronic Heart FailureESC201220 Chronic Heart Failure: Management of Chronic Heart Failure in Adults in Primary and Secondary CareNICE201021 Antithrombotic Therapy and Prevention of ThrombosisACCP201222 Guidelines for the Care of Heart Transplant RecipientsISHLT201023Statements Contemporary Definitions and Classification of the CardiomyopathiesAHA200624 Genetics and Cardiovascular DiseaseAHA201225 Appropriate Utilization of Cardiovascular Imaging in Heart FailureACCF201326 Appropriate Use Criteria for Coronary Revascularization Focused UpdateACCF201227 Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood PressureNHLBI200328 Implications of Recent Clinical Trials for the National Cholesterol Education Program Adult Treatment Panel III GuidelinesNHLBI200229 Referral, Enrollment, and Delivery of Cardiac Rehabilitation/Secondary Prevention Programs at Clinical Centers and BeyondAHA/AACVPR201130 Decision Making in Advanced Heart FailureAHA201231 Recommendations for the Use of Mechanical Circulatory Support: Device Strategies and Patient SelectionAHA201232 Advanced Chronic Heart FailureESC200733 Oral Antithrombotic Agents for the Prevention of Stroke in Nonvalvular Atrial FibrillationAHA/ASA201234 Third Universal Definition of Myocardial InfarctionESC/ACCF/AHA/WHF201235AACVPR indicates American Association of Cardiovascular and Pulmonary Rehabilitation; AATS, American Association for Thoracic Surgery; ACCF, American College of Cardiology Foundation; ACCP, American College of Chest Physicians; ACP, American College of Physicians; AHA, American Heart Association; ASA, American Stroke Association; ESC, European Society of Cardiology; HFSA, Heart Failure Society of America; HRS, Heart Rhythm Society; ISHLT, International Society for Heart and Lung Transplantation; NHLBI, National Heart, Lung, and Blood Institute; NICE, National Institute for Health and Clinical Excellence; PCNA, Preventive Cardiovascular Nurses Association; SCAI, Society for Cardiovascular Angiography and Interventions; STS, Society of Thoracic Surgeons; and WHF, World Heart Federation.2. Definition of HFHF is a complex clinical syndrome that results from any structural or functional impairment of ventricular filling or ejection of blood. The cardinal manifestations of HF are dyspnea and fatigue, which may limit exercise tolerance, and fluid retention, which may lead to pulmonary and/or splanchnic congestion and/or peripheral edema. Some patients have exercise intolerance but little evidence of fluid retention, whereas others complain primarily of edema, dyspnea, or fatigue. Because some patients present without signs or symptoms of volume overload, the term “heart failure” is preferred over “congestive heart failure.” There is no single diagnostic test for HF because it is largely a clinical diagnosis based on a careful history and physical examination.The clinical syndrome of HF may result from disorders of the pericardium, myocardium, endocardium, heart valves, or great vessels, or from certain metabolic abnormalities, but most patients with HF have symptoms due to impaired left ventricular (LV) myocardial function. It should be emphasized that HF is not synonymous with either cardiomyopathy or LV dysfunction; these latter terms describe possible structural or functional reasons for the development of HF. HF may be associated with a wide spectrum of LV functional abnormalities, which may range from patients with normal LV size and preserved EF to those with severe dilatation and/or markedly reduced EF. In most patients, abnormalities of systolic and diastolic dysfunction coexist, irrespective of EF. EF is considered important in classification of patients with HF because of differing patient demographics, comorbid conditions, prognosis, and response to therapies36 and because most clinical trials selected patients based on EF. EF values are dependent on the imaging technique used, method of analysis, and operator. As other techniques may indicate abnormalities in systolic function among patients with a preserved EF, it is preferable to use the terms preserved or reduced EF over preserved or reduced systolic function. For the remainder of this guideline, we will consistently refer to HF with preserved EF and HF with reduced EF as HFpEF and HFrEF, respectively (Table 3).Table 3. Definitions of HFrEF and HFpEFClassificationEF (%)DescriptionI. Heart failure with reduced ejection fraction (HFrEF)≤40Also referred to as systolic HF. Randomized controlled trials have mainly enrolled patients with HFrEF, and it is only in these patients that efficacious therapies have been demonstrated to date.II. Heart failure with preserved ejection fraction (HFpEF)≥50Also referred to as diastolic HF. Several different criteria have been used to further define HFpEF. The diagnosis of HFpEF is challenging because it is largely one of excluding other potential noncardiac causes of symptoms suggestive of HF. To date, efficacious therapies have not been identified. a. HFpEF, borderline41 to 49These patients fall into a borderline or intermediate group. Their characteristics, treatment patterns, and outcomes appear similar to those of patients with HFpEF. b. HFpEF, improved>40It has been recognized that a subset of patients with HFpEF previously had HFrEF. These patients with improvement or recovery in EF may be clinically distinct from those with persistently preserved or reduced EF. Further research is needed to better characterize these patients.EF indicates ejection fraction; HF, heart failure; HFpEF, heart failure with preserved ejection fraction; and HFrEF, heart failure with reduced ejection fraction3. HF ClassificationsBoth the ACCF/AHA stages of HF37 and the New York Heart Association (NYHA) functional classification37,38 provide useful and complementary information about the presence and severity of HF. The ACCF/AHA stages of HF emphasize the development and progression of disease and can be used to describe individuals and populations, whereas the NYHA classes focus on exercise capacity and the symptomatic status of the disease (Table 4).Table 4. Comparison of ACCF/AHA Stages of HF and NYHA Functional ClassificationsACCF/AHA Stages of HF37NYHA Functional Classification38AAt high risk for HF but without structural heart disease or symptoms of HFNoneBStructural heart disease but without signs or symptoms of HFINo limitation of physical activity. Ordinary physical activity does not cause symptoms of HF.CStructural heart disease with prior or current symptoms of HFINo limitation of physical activity. Ordinary physical activity does not cause symptoms of HF.IISlight limitation of physical activity. Comfortable at rest, but ordinary physical
DOI: 10.1056/nejmoa1109400
2013
Cited 2,560 times
Intestinal Microbial Metabolism of Phosphatidylcholine and Cardiovascular Risk
Recent studies in animals have shown a mechanistic link between intestinal microbial metabolism of the choline moiety in dietary phosphatidylcholine (lecithin) and coronary artery disease through the production of a proatherosclerotic metabolite, trimethylamine-N-oxide (TMAO). We investigated the relationship among intestinal microbiota-dependent metabolism of dietary phosphatidylcholine, TMAO levels, and adverse cardiovascular events in humans.
DOI: 10.1161/cir.0b013e31829e8776
2013
Cited 1,727 times
2013 ACCF/AHA Guideline for the Management of Heart Failure
HomeCirculationVol. 128, No. 162013 ACCF/AHA Guideline for the Management of Heart Failure Free AccessResearch ArticlePDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissionsDownload Articles + Supplements ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toSupplemental MaterialFree AccessResearch ArticlePDF/EPUB2013 ACCF/AHA Guideline for the Management of Heart FailureA Report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines , Clyde W. Yancy, MD, MSc, FACC, FAHA, Chair, Mariell Jessup, MD, FACC, FAHA, Vice Chair, Biykem Bozkurt, MD, PhD, FACC, FAHA, Javed Butler, MBBS, FACC, FAHA, Donald E. CaseyJr, MD, MPH, MBA, FACP, FAHA, Mark H. Drazner, MD, MSc, FACC, FAHA, Gregg C. Fonarow, MD, FACC, FAHA, Stephen A. Geraci, MD, FACC, FAHA, FCCP, Tamara Horwich, MD, FACC, James L. Januzzi, MD, FACC, Maryl R. Johnson, MD, FACC, FAHA, Edward K. Kasper, MD, FACC, FAHA, Wayne C. Levy, MD, FACC, Frederick A. Masoudi, MD, MSPH, FACC, FAHA, Patrick E. McBride, MD, MPH, FACC, John J.V. McMurray, MD, FACC, Judith E. Mitchell, MD, FACC, FAHA, Pamela N. Peterson, MD, MSPH, FACC, FAHA, Barbara Riegel, DNSc, RN, FAHA, Flora Sam, MD, FACC, FAHA, Lynne W. Stevenson, MD, FACC, W.H. Wilson Tang, MD, FACC, Emily J. Tsai, MD, FACC and Bruce L. Wilkoff, MD, FACC, FHRS Search for more papers by this author , Clyde W. YancyClyde W. Yancy Search for more papers by this author , Mariell JessupMariell Jessup Search for more papers by this author , Biykem BozkurtBiykem Bozkurt Search for more papers by this author , Javed ButlerJaved Butler Search for more papers by this author , Donald E. CaseyJrDonald E. CaseyJr Search for more papers by this author , Mark H. DraznerMark H. Drazner Search for more papers by this author , Gregg C. FonarowGregg C. Fonarow Search for more papers by this author , Stephen A. GeraciStephen A. Geraci Search for more papers by this author , Tamara HorwichTamara Horwich Search for more papers by this author , James L. JanuzziJames L. Januzzi Search for more papers by this author , Maryl R. JohnsonMaryl R. Johnson Search for more papers by this author , Edward K. KasperEdward K. Kasper Search for more papers by this author , Wayne C. LevyWayne C. Levy Search for more papers by this author , Frederick A. MasoudiFrederick A. Masoudi Search for more papers by this author , Patrick E. McBridePatrick E. McBride Search for more papers by this author , John J.V. McMurrayJohn J.V. McMurray Search for more papers by this author , Judith E. MitchellJudith E. Mitchell Search for more papers by this author , Pamela N. PetersonPamela N. Peterson Search for more papers by this author , Barbara RiegelBarbara Riegel Search for more papers by this author , Flora SamFlora Sam Search for more papers by this author , Lynne W. StevensonLynne W. Stevenson Search for more papers by this author , W.H. Wilson TangW.H. Wilson Tang Search for more papers by this author , Emily J. TsaiEmily J. Tsai Search for more papers by this author and Bruce L. WilkoffBruce L. Wilkoff Search for more papers by this author and WRITING COMMITTEE MEMBERS Originally published5 Jun 2013https://doi.org/10.1161/CIR.0b013e31829e8776Circulation. 2013;128:e240–e327Other version(s) of this articleYou are viewing the most recent version of this article. Previous versions: January 1, 2013: Previous Version 1 Table of ContentsPreamble e2421. Introduction e2451.1. Methodology and Evidence Review e2451.2. Organization of the Writing Committee e2451.3. Document Review and Approval e2451.4. Scope of This Guideline With Reference to Other Relevant Guidelines or Statements e2452. Definition of HF e2462.1. HF With Reduced EF (HFrEF) e2472.2. HF With Preserved EF (HFpEF) e2473. HF Classifications e2474. Epidemiology e2484.1. Mortality e2484.2. Hospitalizations e2484.3. Asymptomatic LV Dysfunction e2484.4. Health-Related Quality of Life and Functional Status e2494.5. Economic Burden of HF e2494.6. Important Risk Factors for HF (Hypertension, Diabetes Mellitus, Metabolic Syndrome, and Atherosclerotic Disease) e2495. Cardiac Structural Abnormalities and Other Causes of HF e2495.1. Dilated Cardiomyopathies e2495.1.1. Definition and Classification of Dilated Cardiomyopathies e2495.1.2. Epidemiology and Natural History of DCM e2505.2. Familial Cardiomyopathies e2505.3. Endocrine and Metabolic Causes of Cardiomyopathy e2505.3.1. Obesity e2505.3.2. Diabetic Cardiomyopathy e2505.3.3. Thyroid Disease e2505.3.4. Acromegaly and Growth Hormone Deficiency e2505.4. Toxic Cardiomyopathy e2515.4.1. Alcoholic Cardiomyopathy e2515.4.2. Cocaine Cardiomyopathy e2515.4.3. Cardiotoxicity Related to Cancer Therapies e2515.4.4. Other Myocardial Toxins and Nutritional Causes of Cardiomyopathy e2515.5. Tachycardia-Induced Cardiomyopathy e2515.6. Myocarditis and Cardiomyopathies Due to Inflammation e2515.6.1. Myocarditis e2515.6.2. Acquired Immunodeficiency Syndrome e2525.6.3. Chagas Disease e2525.7. Inflammation-Induced Cardiomyopathy: Noninfectious Causes e2525.7.1. Hypersensitivity Myocarditis e2525.7.2. Rheumatological/Connective Tissue Disorders e2525.8. Peripartum Cardiomyopathy e2525.9. Cardiomyopathy Caused By Iron Overload e2525.10. Amyloidosis e2525.11. Cardiac Sarcoidosis e2535.12. Stress (Takotsubo) Cardiomyopathy e2536. Initial and Serial Evaluation of the HF Patient e2536.1. Clinical Evaluation e2536.1.1. History and Physical Examination: Recommendations e2536.1.2. Risk Scoring: Recommendation e2536.2. Diagnostic Tests: Recommendations e2536.3. Biomarkers: Recommendations e2556.3.1. Natriuretic Peptides: BNP or NT-proBNP e2566.3.2. Biomarkers of Myocardial Injury: Cardiac Troponin T or I e2566.3.3. Other Emerging Biomarkers e2566.4. Noninvasive Cardiac Imaging: Recommendations e2566.5. Invasive Evaluation: Recommendations e2586.5.1. Right-Heart Catheterization e2596.5.2. Left-Heart Catheterization e2596.5.3. Endomyocardial Biopsy e2607. Treatment of Stages A to D e2607.1. Stage A: Recommendations e2607.1.1. Recognition and Treatment of Elevated Blood Pressure e2607.1.2. Treatment of Dyslipidemia and Vascular Risk e2607.1.3. Obesity and Diabetes Mellitus e2607.1.4. Recognition and Control of Other Conditions That May Lead to HF e2607.2. Stage B: Recommendations e2617.2.1. Management Strategies for Stage B e2627.3. Stage C e2627.3.1. Nonpharmacological Interventions e2627.3.1.1. Education: Recommendation e2627.3.1.2. Social Support e2637.3.1.3. Sodium Restriction: Recommendation e2637.3.1.4. Treatment of Sleep Disorders: Recommendation e2637.3.1.5. Weight Loss e2637.3.1.6. Activity, Exercise Prescription, and Cardiac Rehabilitation: Recommendations e2647.3.2. Pharmacological Treatment for Stage C HFrEF: Recommendations e2647.3.2.1. Diuretics: Recommendation e2657.3.2.2. ACE Inhibitors: Recommendation e2657.3.2.3. ARBs: Recommendations e2677.3.2.4. Beta Blockers: Recommendation e2677.3.2.5. Aldosterone Receptor Antagonists: Recommendations e2687.3.2.6. Hydralazine and Isosorbide Dinitrate: Recommendations e2707.3.2.7. Digoxin: Recommendation e2717.3.2.8. Other Drug Treatment e2717.3.2.8.1. Anticoagulation: Recommendations e2717.3.2.8.2. Statins: Recommendation e2727.3.2.8.3. Omega-3 Fatty Acids: Recommendation e2727.3.2.9. Drugs of Unproven Value or That May Worsen HF: Recommendations e2737.3.2.9.1. Nutritional Supplements and Hormonal Therapies e2737.3.2.9.2. Antiarrhythmic Agents e2737.3.2.9.3. Calcium Channel Blockers: Recommendation e2737.3.2.9.4. Nonsteroidal Anti-Inflammatory Drugs e2747.3.2.9.5. Thiazolidinediones e2747.3.3. Pharmacological Treatment for Stage C HFpEF: Recommendations e2747.3.4. Device Therapy for Stage C HFrEF: Recommendations e2747.3.4.1. Implantable Cardioverter-Defibrillator e2787.3.4.2. Cardiac Resynchronization Therapy e2797.4. Stage D e2807.4.1. Definition of Advanced HF e2807.4.2. Important Considerations in Determining If the Patient Is Refractory e2807.4.3. Water Restriction: Recommendation e2807.4.4. Inotropic Support: Recommendations e2817.4.5. Mechanical Circulatory Support: Recommendations e2827.4.6. Cardiac Transplantation: Recommendation e2838. The Hospitalized Patient e2848.1. Classification of Acute Decompensated HF e2848.2. Precipitating Causes of Decompensated HF: Recommendations e2858.3. Maintenance of GDMT During Hospitalization: Recommendations e2868.4. Diuretics in Hospitalized Patients: Recommendations e2868.5. Renal Replacement Therapy—Ultrafiltration: Recommendations e2878.6. Parenteral Therapy in Hospitalized HF: Recommendation e2878.7. Venous Thromboembolism Prophylaxis in Hospitalized Patients: Recommendation e2888.8. Arginine Vasopressin Antagonists: Recommendation e2888.9. Inpatient and Transitions of Care: Recommendations e2889. Important Comorbidities in HF e2909.1. Atrial Fibrillation e2909.2. Anemia e2939.3. Depression e2939.4. Other Multiple Comorbidities e29310. Surgical/Percutaneous/Transcatheter Interventional Treatments of HF: Recommendations e29311. Coordinating Care for Patients With Chronic HF e29511.1. Coordinating Care for Patients With Chronic HF: Recommendations e29511.2. Systems of Care to Promote Care Coordination for Patients With Chronic HF e29611.3. Palliative Care for Patients With HF e29612. Quality Metrics/Performance Measures: Recommendations e29613. Evidence Gaps and Future Research Directions e299References e299Appendix 1. Author Relationships With Industry and Other Entities (Relevant) e320Appendix 2. Reviewer Relationships With Industry and Other Entities (Relevant) e323Appendix 3. Abbreviations e327PreambleThe medical profession should play a central role in evaluating the evidence related to drugs, devices, and procedures for the detection, management, and prevention of disease. When properly applied, expert analysis of available data on the benefits and risks of these therapies and procedures can improve the quality of care, optimize patient outcomes, and favorably affect costs by focusing resources on the most effective strategies. An organized and directed approach to a thorough review of evidence has resulted in the production of clinical practice guidelines that assist clinicians in selecting the best management strategy for an individual patient. Moreover, clinical practice guidelines can provide a foundation for other applications, such as performance measures, appropriate use criteria, and both quality improvement and clinical decision support tools.The American College of Cardiology Foundation (ACCF) and the American Heart Association (AHA) have jointly produced guidelines in the area of cardiovascular disease since 1980. The ACCF/AHA Task Force on Practice Guidelines (Task Force), charged with developing, updating, and revising practice guidelines for cardiovascular diseases and procedures, directs and oversees this effort. Writing committees are charged with regularly reviewing and evaluating all available evidence to develop balanced, patient-centric recommendations for clinical practice.Experts in the subject under consideration are selected by the ACCF and AHA to examine subject-specific data and write guidelines in partnership with representatives from other medical organizations and specialty groups. Writing committees are asked to perform a literature review; weigh the strength of evidence for or against particular tests, treatments, or procedures; and include estimates of expected outcomes where such data exist. Patient-specific modifiers, comorbidities, and issues of patient preference that may influence the choice of tests or therapies are considered. When available, information from studies on cost is considered, but data on efficacy and outcomes constitute the primary basis for the recommendations contained herein.In analyzing the data and developing recommendations and supporting text, the writing committee uses evidence-based methodologies developed by the Task Force.1 The Class of Recommendation (COR) is an estimate of the size of the treatment effect considering risks versus benefits in addition to evidence and/or agreement that a given treatment or procedure is or is not useful/effective or in some situations may cause harm. The Level of Evidence (LOE) is an estimate of the certainty or precision of the treatment effect. The writing committee reviews and ranks evidence supporting each recommendation with the weight of evidence ranked as LOE A, B, or C according to specific definitions that are included in Table 1. Studies are identified as observational, retrospective, prospective, or randomized where appropriate. For certain conditions for which inadequate data are available, recommendations are based on expert consensus and clinical experience and are ranked as LOE C. When recommendations at LOE C are supported by historical clinical data, appropriate references (including clinical reviews) are cited if available. For issues for which sparse data are available, a survey of current practice among the clinicians on the writing committee is the basis for LOE C recommendations and no references are cited. The schema for COR and LOE are summarized in Table 1, which also provides suggested phrases for writing recommendations within each COR. A new addition to this methodology is separation of the Class III recommendations to delineate whether the recommendation is determined to be of “no benefit” or is associated with “harm” to the patient. In addition, in view of the increasing number of comparative effectiveness studies, comparator verbs and suggested phrases for writing recommendations for the comparative effectiveness of one treatment or strategy versus another have been added for COR I and IIa, LOE A or B only.Table 1. Applying Classification of Recommendation and Level of EvidenceTable 1. Applying Classification of Recommendation and Level of EvidenceIn view of the advances in medical therapy across the spectrum of cardiovascular diseases, the Task Force has designated the term guideline-directed medical therapy (GDMT) to represent optimal medical therapy as defined by ACCF/AHA guideline–recommended therapies (primarily Class I). This new term, GDMT, will be used herein and throughout all future guidelines.Because the ACCF/AHA practice guidelines address patient populations (and clinicians) residing in North America, drugs that are not currently available in North America are discussed in the text without a specific COR. For studies performed in large numbers of subjects outside North America, each writing committee reviews the potential influence of different practice patterns and patient populations on the treatment effect and relevance to the ACCF/AHA target population to determine whether the findings should inform a specific recommendation.The ACCF/AHA practice guidelines are intended to assist clinicians in clinical decision making by describing a range of generally acceptable approaches to the diagnosis, management, and prevention of specific diseases or conditions. The guidelines attempt to define practices that meet the needs of most patients in most circumstances. The ultimate judgment regarding care of a particular patient must be made by the clinician and patient in light of all the circumstances presented by that patient. As a result, situations may arise for which deviations from these guidelines may be appropriate. Clinical decision making should involve consideration of the quality and availability of expertise in the area where care is provided. When these guidelines are used as the basis for regulatory or payer decisions, the goal should be improvement in quality of care. The Task Force recognizes that situations arise in which additional data are needed to inform patient care more effectively; these areas will be identified within each respective guideline when appropriate.Prescribed courses of treatment in accordance with these recommendations are effective only if followed. Because lack of patient understanding and adherence may adversely affect outcomes, clinicians should make every effort to engage the patient’s active participation in prescribed medical regimens and lifestyles. In addition, patients should be informed of the risks, benefits, and alternatives to a particular treatment and be involved in shared decision making whenever feasible, particularly for COR IIa and IIb, for which the benefit-to-risk ratio may be lower.The Task Force makes every effort to avoid actual, potential, or perceived conflicts of interest that may arise as a result of industry relationships or personal interests among the members of the writing committee. All writing committee members and peer reviewers of the guideline are required to disclose all current healthcare-related relationships, including those existing 12 months before initiation of the writing effort. In December 2009, the ACCF and AHA implemented a new policy for relationship with industry and other entities (RWI) that requires the writing committee chair plus a minimum of 50% of the writing committee to have no relevant RWI (Appendix 1 includes the ACCF/AHA definition of relevance). These statements are reviewed by the Task Force and all members during each conference call and/or meeting of the writing committee and are updated as changes occur. All guideline recommendations require a confidential vote by the writing committee and must be approved by a consensus of the voting members. Members are not permitted to draft or vote on any text or recommendations pertaining to their RWI. Members who recused themselves from voting are indicated in the list of writing committee members, and specific section recusals are noted in Appendix 1. Authors’ and peer reviewers’ RWI pertinent to this guideline are disclosed in Appendixes 1 and 2, respectively. Additionally, to ensure complete transparency, writing committee members’ comprehensive disclosure information—including RWI not pertinent to this document—is available as an online supplement. Comprehensive disclosure information for the Task Force is also available online at http://www.cardiosource.org/en/ACC/About-ACC/Who-We-Are/Leadership/Guidelines-and-Documents-Task-Forces.aspx. The work of writing committees is supported exclusively by the ACCF and AHA without commercial support. Writing committee members volunteered their time for this activity.In an effort to maintain relevance at the point of care for practicing clinicians, the Task Force continues to oversee an ongoing process improvement initiative. As a result, in response to pilot projects, several changes to these guidelines will be apparent, including limited narrative text, a focus on summary and evidence tables (with references linked to abstracts in PubMed), and more liberal use of summary recommendation tables (with references that support LOE) to serve as a quick reference.In April 2011, the Institute of Medicine released 2 reports: Clinical Practice Guidelines We Can Trust and Finding What Works in Health Care: Standards for Systematic Reviews.2,3 It is noteworthy that the ACCF/AHA practice guidelines are cited as being compliant with many of the proposed standards. A thorough review of these reports and of our current methodology is under way, with further enhancements anticipated.The recommendations in this guideline are considered current until they are superseded by a focused update or the full-text guideline is revised. Guidelines are official policy of both the ACCF and AHA.Jeffrey L. Anderson, MD, FACC, FAHAChair, ACCF/AHA Task Force on Practice Guidelines1. Introduction1.1. Methodology and Evidence ReviewThe recommendations listed in this document are, whenever possible, evidence based. An extensive evidence review was conducted through October 2011 and includes selected other references through April 2013. Searches were extended to studies, reviews, and other evidence conducted in human subjects and that were published in English from PubMed, EMBASE, Cochrane, Agency for Healthcare Research and Quality Reports, and other selected databases relevant to this guideline. Key search words included but were not limited to the following: heart failure, cardiomyopathy, quality of life, mortality, hospitalizations, prevention, biomarkers, hypertension, dyslipidemia, imaging, cardiac catheterization, endomyocardial biopsy, angiotensin-converting enzyme inhibitors, angiotensin-receptor antagonists/blockers, beta blockers, cardiac, cardiac resynchronization therapy, defibrillator, device-based therapy, implantable cardioverter-defibrillator, device implantation, medical therapy, acute decompensated heart failure, preserved ejection fraction, terminal care and transplantation, quality measures, and performance measures. Additionally, the committee reviewed documents related to the subject matter previously published by the ACCF and AHA. References selected and published in this document are representative and not all-inclusive.To provide clinicians with a representative evidence base, whenever deemed appropriate or when published, the absolute risk difference and number needed to treat or harm are provided in the guideline (within tables), along with confidence intervals and data related to the relative treatment effects such as odds ratio, relative risk, hazard ratio, and incidence rate ratio.1.2. Organization of the Writing CommitteeThe committee was composed of physicians and a nurse with broad expertise in the evaluation, care, and management of patients with heart failure (HF). The authors included general cardiologists, HF and transplant specialists, electrophysiologists, general internists, and physicians with methodological expertise. The committee included representatives from the ACCF, AHA, American Academy of Family Physicians, American College of Chest Physicians, American College of Physicians, Heart Rhythm Society, and International Society for Heart and Lung Transplantation.1.3. Document Review and ApprovalThis document was reviewed by 2 official reviewers each nominated by both the ACCF and the AHA, as well as 1 to 2 reviewers each from the American Academy of Family Physicians, American College of Chest Physicians, Heart Rhythm Society, and International Society for Heart and Lung Transplantation, as well as 32 individual content reviewers (including members of the ACCF Adult Congenital and Pediatric Cardiology Council, ACCF Cardiovascular Team Council, ACCF Council on Cardiovascular Care for Older Adults, ACCF Electrophysiology Committee, ACCF Heart Failure and Transplant Council, ACCF Imaging Council, ACCF Prevention Committee, ACCF Surgeons’ Scientific Council, and ACCF Task Force on Appropriate Use Criteria). All information on reviewers’ RWI was distributed to the writing committee and is published in this document (Appendix 2).This document was approved for publication by the governing bodies of the ACCF and AHA and endorsed by the American Association of Cardiovascular and Pulmonary Rehabilitation, American College of Chest Physicians, Heart Rhythm Society, and International Society for Heart and Lung Transplantation.1.4. Scope of This Guideline With Reference to Other Relevant Guidelines or StatementsThis guideline covers multiple management issues for the adult patient with HF. Although there is an abundance of evidence addressing HF, for many important clinical considerations, this writing committee was unable to identify sufficient data to properly inform a recommendation. The writing committee actively worked to reduce the number of LOE “C” recommendations, especially for Class I−recommended therapies. Despite these limitations, it is apparent that much can be done for HF. Adherence to the clinical practice guidelines herein reproduced should lead to improved patient outcomes.Although of increasing importance, HF in children and congenital heart lesions in adults are not specifically addressed in this guideline. The reader is referred to publically available resources to address questions in these areas. However, this guideline does address HF with preserved ejection fraction (EF) in more detail and similarly revisits hospitalized HF. Additional areas of renewed interest are in stage D HF, palliative care, transition of care, and quality of care for HF. Certain management strategies appropriate for the patient at risk for HF or already affected by HF are also reviewed in numerous relevant clinical practice guidelines and scientific statements published by the ACCF/AHA Task Force on Practice Guidelines, AHA, ACCF Task Force on Appropriate Use Criteria, European Society of Cardiology, Heart Failure Society of America, and the National Heart, Lung, and Blood Institute. The writing committee saw no need to reiterate the recommendations contained in those guidelines and chose to harmonize recommendations when appropriate and eliminate discrepancies. This is especially the case for device-based therapeutics, where complete alignment between the HF guideline and the device-based therapy guideline was deemed imperative.4 Some recommendations from earlier guidelines have been updated as warranted by new evidence or a better understanding of earlier evidence, whereas others that were no longer accurate or relevant or which were overlapping were modified; recommendations from previous guidelines that were similar or redundant were eliminated or consolidated when possible.The present document recommends a combination of lifestyle modifications and medications that constitute GDMT. GDMT is specifically referenced in the recommendations for the treatment of HF (Section 7.3.2). Both for GDMT and other recommended drug treatment regimens, the reader is advised to confirm dosages with product insert material and to evaluate carefully for contraindications and drug-drug interactions. Table 2 is a list of documents deemed pertinent to this effort and is intended for use as a resource; it obviates the need to repeat already extant guideline recommendations. Additional other HF guideline statements are highlighted as well for the purpose of comparison and completeness.Table 2. Associated Guidelines and StatementsTitleOrganizationPublication Year (Reference)Guidelines Guidelines for the Management of Adults With Congenital Heart DiseaseACCF/AHA20085 Guidelines for the Management of Patients With Atrial FibrillationACCF/AHA/HRS20116–8 Guideline for Assessment of Cardiovascular Risk in Asymptomatic AdultsACCF/AHA20109 Guideline for Coronary Artery Bypass Graft SurgeryACCF/AHA201110 Guidelines for Device-Based Therapy of Cardiac Rhythm AbnormalitiesACCF/AHA/HRS20134 Guideline for the Diagnosis and Treatment of Hypertrophic CardiomyopathyACCF/AHA201111 Guideline for Percutaneous Coronary InterventionACCF/AHA/SCAI201112 Secondary Prevention and Risk Reduction Therapy for Patients With Coronary and Other Atherosclerotic Vascular Disease: 2011 UpdateAHA/ACCF201113 Guideline for the Diagnosis and Management of Patients With Stable Ischemic Heart DiseaseACCF/AHA/ACP/AATS/PCNA/SCAI/STS201214 Guideline for the Management of ST-Elevation Myocardial InfarctionACCF/AHA201315 Guidelines for the Management of Patients With Unstable Angina/Non–ST-Elevation Myocardial InfarctionACCF/AHA201316 Guidelines for the Management of Patients With Valvular Heart DiseaseACCF/AHA200817 Comprehensive Heart Failure Practice GuidelineHFSA201018 Guidelines for the Diagnosis and Treatment of Acute and Chronic Heart FailureESC201219 Chronic Heart Failure: Management of Chronic Heart Failure in Adults in Primary and Secondary CareNICE201020 Antithrombotic Therapy and Prevention of ThrombosisACCP201221 Guidelines for the Care of Heart Transplant RecipientsISHLT201022Statements Contemporary Definitions and Classification of the CardiomyopathiesAHA200623 Genetics and Cardiovascular DiseaseAHA201224 Appropriate Utilization of Cardiovascular Imaging in Heart FailureACCF201325 Appropriate Use Criteria for Coronary Revascularization Focused UpdateACCF201226 Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood PressureNHLBI200327 Implications of Recent Clinical Trials for the National Cholesterol Education Program Adult Treatment Panel III GuidelinesNHLBI200228 Referral, Enrollment, and Delivery of Cardiac Rehabilitation/Secondary Prevention Programs at Clinical Centers and BeyondAHA/AACVPR201129 Decision Making in Advanced Heart FailureAHA201230 Recommendations for the Use of Mechanical Circulatory Support: Device Strategies and Patient SelectionAHA201231 Advanced Chronic Heart FailureESC200732 Oral Antithrombotic Agents for the Prevention of Stroke in Nonvalvular Atrial FibrillationAHA/ASA201233 Third Universal Definition of Myocardial InfarctionESC/ACCF/AHA/WHF201234AACVPR indicates American Association of Cardiovascular and Pulmonary Rehabilitation; AATS, American Association for Thoracic Surgery; ACCF, American College of Cardiology Foundation; ACCP, American College of Chest Physicians; ACP, American College of Physicians; AHA, American Heart Association; ASA, American Stroke Association; ESC, European Society of Cardiology; HFSA, Heart Failure Society of America; HRS, Heart Rhythm Society; ISHLT, International Society for Heart and Lung Transplantation; NHLBI, National Heart, Lung, and Blood Institute; NICE, National Institute for Health and Clinical Excellence; PCNA, Preventive Cardiovascular Nurses Association; SCAI, Society for Cardiovascular Angiography and Interventions; STS, Society of Thoracic Surgeons; and WHF, World Heart Federation.2. Definition of HFHF is a complex clinical syndrome that results from any structural or functional impairment of ventricular filling or ejection of blood. The cardinal manifestations of HF are dyspnea and fatigue, which may limit exercise tolerance, a
DOI: 10.1016/j.cell.2016.02.011
2016
Cited 1,357 times
Gut Microbial Metabolite TMAO Enhances Platelet Hyperreactivity and Thrombosis Risk
<h2>Summary</h2> Normal platelet function is critical to blood hemostasis and maintenance of a closed circulatory system. Heightened platelet reactivity, however, is associated with cardiometabolic diseases and enhanced potential for thrombotic events. We now show gut microbes, through generation of trimethylamine N-oxide (TMAO), directly contribute to platelet hyperreactivity and enhanced thrombosis potential. Plasma TMAO levels in subjects (n > 4,000) independently predicted incident (3 years) thrombosis (heart attack, stroke) risk. Direct exposure of platelets to TMAO enhanced sub-maximal stimulus-dependent platelet activation from multiple agonists through augmented Ca<sup>2+</sup> release from intracellular stores. Animal model studies employing dietary choline or TMAO, germ-free mice, and microbial transplantation collectively confirm a role for gut microbiota and TMAO in modulating platelet hyperresponsiveness and thrombosis potential and identify microbial taxa associated with plasma TMAO and thrombosis potential. Collectively, the present results reveal a previously unrecognized mechanistic link between specific dietary nutrients, gut microbes, platelet function, and thrombosis risk.
DOI: 10.1016/j.jacc.2008.05.068
2009
Cited 1,340 times
Importance of Venous Congestion for Worsening of Renal Function in Advanced Decompensated Heart Failure
To determine whether venous congestion, rather than impairment of cardiac output, is primarily associated with the development of worsening renal function (WRF) in patients with advanced decompensated heart failure (ADHF). Reduced cardiac output is traditionally believed to be the main determinant of WRF in patients with ADHF. A total of 145 consecutive patients admitted with ADHF treated with intensive medical therapy guided by pulmonary artery catheter were studied. We defined WRF as an increase of serum creatinine ≥0.3 mg/dl during hospitalization. In the study cohort (age 57 ± 14 years, cardiac index 1.9 ± 0.6 l/min/m2, left ventricular ejection fraction 20 ± 8%, serum creatinine 1.7 ± 0.9 mg/dl), 58 patients (40%) developed WRF. Patients who developed WRF had a greater central venous pressure (CVP) on admission (18 ± 7 mm Hg vs. 12 ± 6 mm Hg, p < 0.001) and after intensive medical therapy (11 ± 8 mm Hg vs. 8 ± 5 mm Hg, p = 0.04). The development of WRF occurred less frequently in patients who achieved a CVP <8 mm Hg (p = 0.01). Furthermore, the ability of CVP to stratify risk for development of WRF was apparent across the spectrum of systemic blood pressure, pulmonary capillary wedge pressure, cardiac index, and estimated glomerular filtration rates. Venous congestion is the most important hemodynamic factor driving WRF in decompensated patients with advanced heart failure.
DOI: 10.1161/circresaha.117.309715
2017
Cited 1,091 times
Gut Microbiota in Cardiovascular Health and Disease
Significant interest in recent years has focused on gut microbiota–host interaction because accumulating evidence has revealed that intestinal microbiota play an important role in human health and disease, including cardiovascular diseases. Changes in the composition of gut microbiota associated with disease, referred to as dysbiosis, have been linked to pathologies such as atherosclerosis, hypertension, heart failure, chronic kidney disease, obesity, and type 2 diabetes mellitus. In addition to alterations in gut microbiota composition, the metabolic potential of gut microbiota has been identified as a contributing factor in the development of diseases. Recent studies revealed that gut microbiota can elicit a variety of effects on the host. Indeed, the gut microbiome functions like an endocrine organ, generating bioactive metabolites, that can impact host physiology. Microbiota interact with the host through many pathways, including the trimethylamine/trimethylamine N -oxide pathway, short-chain fatty acids pathway, and primary and secondary bile acids pathways. In addition to these metabolism-dependent pathways, metabolism-independent processes are suggested to also potentially contribute to cardiovascular disease pathogenesis. For example, heart failure–associated splanchnic circulation congestion, bowel wall edema, and impaired intestinal barrier function are thought to result in bacterial translocation, the presence of bacterial products in the systemic circulation and heightened inflammatory state. These are thought to also contribute to further progression of heart failure and atherosclerosis. The purpose of the current review is to highlight the complex interplay between microbiota, their metabolites, and the development and progression of cardiovascular diseases. We will also discuss the roles of gut microbiota in normal physiology and the potential of modulating intestinal microbial inhabitants as novel therapeutic targets.
DOI: 10.1161/circresaha.116.305360
2015
Cited 895 times
Gut Microbiota-Dependent Trimethylamine <i>N</i> -Oxide (TMAO) Pathway Contributes to Both Development of Renal Insufficiency and Mortality Risk in Chronic Kidney Disease
Rationale: Trimethylamine- N -oxide (TMAO), a gut microbial-dependent metabolite of dietary choline, phosphatidylcholine (lecithin), and l -carnitine, is elevated in chronic kidney diseases (CKD) and associated with coronary artery disease pathogenesis. Objective: To both investigate the clinical prognostic value of TMAO in subjects with versus without CKD, and test the hypothesis that TMAO plays a direct contributory role in the development and progression of renal dysfunction. Methods and Results: We first examined the relationship between fasting plasma TMAO and all-cause mortality over 5-year follow-up in 521 stable subjects with CKD (estimated glomerular filtration rate, &lt;60 mL/min per 1.73 m 2 ). Median TMAO level among CKD subjects was 7.9 μmol/L (interquartile range, 5.2–12.4 μmol/L), which was markedly higher ( P &lt;0.001) than in non-CKD subjects (n=3166). Within CKD subjects, higher (fourth versus first quartile) plasma TMAO level was associated with a 2.8-fold increased mortality risk. After adjustments for traditional risk factors, high-sensitivity C-reactive protein, estimated glomerular filtration rate, elevated TMAO levels remained predictive of 5-year mortality risk (hazard ratio, 1.93; 95% confidence interval, 1.13–3.29; P &lt;0.05). TMAO provided significant incremental prognostic value (net reclassification index, 17.26%; P &lt;0.001 and differences in area under receiver operator characteristic curve, 63.26% versus 65.95%; P =0.036). Among non-CKD subjects, elevated TMAO levels portend poorer prognosis within cohorts of high and low cystatin C. In animal models, elevated dietary choline or TMAO directly led to progressive renal tubulointerstitial fibrosis and dysfunction. Conclusions: Plasma TMAO levels are both elevated in patients with CKD and portend poorer long-term survival. Chronic dietary exposures that increase TMAO directly contributes to progressive renal fibrosis and dysfunction in animal models.
DOI: 10.1161/cir.0000000000000664
2019
Cited 653 times
Cardiorenal Syndrome: Classification, Pathophysiology, Diagnosis, and Treatment Strategies: A Scientific Statement From the American Heart Association
Cardiorenal syndrome encompasses a spectrum of disorders involving both the heart and kidneys in which acute or chronic dysfunction in 1 organ may induce acute or chronic dysfunction in the other organ. It represents the confluence of heart-kidney interactions across several interfaces. These include the hemodynamic cross-talk between the failing heart and the response of the kidneys and vice versa, as well as alterations in neurohormonal markers and inflammatory molecular signatures characteristic of its clinical phenotypes. The mission of this scientific statement is to describe the epidemiology and pathogenesis of cardiorenal syndrome in the context of the continuously evolving nature of its clinicopathological description over the past decade. It also describes diagnostic and therapeutic strategies applicable to cardiorenal syndrome, summarizes cardiac-kidney interactions in special populations such as patients with diabetes mellitus and kidney transplant recipients, and emphasizes the role of palliative care in patients with cardiorenal syndrome. Finally, it outlines the need for a cardiorenal education track that will guide future cardiorenal trials and integrate the clinical and research needs of this important field in the future.
DOI: 10.1002/ejhf.1369
2019
Cited 639 times
The use of diuretics in heart failure with congestion — a position statement from the Heart Failure Association of the European Society of Cardiology
The vast majority of acute heart failure episodes are characterized by increasing symptoms and signs of congestion with volume overload. The goal of therapy in those patients is the relief of congestion through achieving a state of euvolaemia, mainly through the use of diuretic therapy. The appropriate use of diuretics however remains challenging, especially when worsening renal function, diuretic resistance and electrolyte disturbances occur. This position paper focuses on the use of diuretics in heart failure with congestion. The manuscript addresses frequently encountered challenges, such as (i) evaluation of congestion and clinical euvolaemia, (ii) assessment of diuretic response/resistance in the treatment of acute heart failure, (iii) an approach towards stepped pharmacologic diuretic strategies, based upon diuretic response, and (iv) management of common electrolyte disturbances. Recommendations are made in line with available guidelines, evidence and expert opinion.
DOI: 10.1016/j.jacc.2014.02.617
2014
Cited 535 times
Prognostic Value of Elevated Levels of Intestinal Microbe-Generated Metabolite Trimethylamine-N-Oxide in Patients With Heart Failure
Altered intestinal function is prevalent in patients with heart failure (HF), but its role in adverse outcomes is unclear.This study investigated the potential pathophysiological contributions of intestinal microbiota in HF.We examined the relationship between fasting plasma trimethylamine-N-oxide (TMAO) and all-cause mortality over a 5-year follow-up in 720 patients with stable HF.The median TMAO level was 5.0 μM, which was higher than in subjects without HF (3.5 μM; p < 0.001). There was modest but significant correlation between TMAO concentrations and B-type natriuretic peptide (BNP) levels (r = 0.23; p < 0.001). Higher plasma TMAO levels were associated with a 3.4-fold increased mortality risk. Following adjustments for traditional risk factors and BNP levels, elevated TMAO levels remained predictive of 5-year mortality risk (hazard ratio [HR]: 2.2; 95% CI: 1.42 to 3.43; p < 0.001), as well as following the addition of estimated glomerular filtration rate to the model (HR: 1.75; 95% CI: 1.07 to 2.86; p < 0.001).High TMAO levels were observed in patients with HF, and elevated TMAO levels portended higher long-term mortality risk independent of traditional risk factors and cardiorenal indexes.
DOI: 10.1172/jci72331
2014
Cited 522 times
The contributory role of gut microbiota in cardiovascular disease
Our group recently discovered that certain dietary nutrients possessing a trimethylamine (TMA) moiety, namely choline/phosphatidylcholine and L-carnitine, participate in the development of atherosclerotic heart disease. A meta-organismal pathway was elucidated involving gut microbiota-dependent formation of TMA and host hepatic flavin monooxygenase 3-dependent (FMO3-dependent) formation of TMA-N-oxide (TMAO), a metabolite shown to be both mechanistically linked to atherosclerosis and whose levels are strongly linked to cardiovascular disease (CVD) risks. Collectively, these studies reveal that nutrient precursors, gut microbiota, and host participants along the meta-organismal pathway elucidated may serve as new targets for the prevention and treatment of CVD.
DOI: 10.1093/eurheartj/ehu002
2014
Cited 468 times
Prognostic value of choline and betaine depends on intestinal microbiota-generated metabolite trimethylamine-N-oxide
Recent metabolomics and animal model studies show trimethylamine-N-oxide (TMAO), an intestinal microbiota-dependent metabolite formed from dietary trimethylamine-containing nutrients such as phosphatidylcholine (PC), choline, and carnitine, is linked to coronary artery disease pathogenesis. Our aim was to examine the prognostic value of systemic choline and betaine levels in stable cardiac patients.We examined the relationship between fasting plasma choline and betaine levels and risk of major adverse cardiac events (MACE = death, myocardial infraction, stroke) in relation to TMAO over 3 years of follow-up in 3903 sequential stable subjects undergoing elective diagnostic coronary angiography. In our study cohort, median (IQR) TMAO, choline, and betaine levels were 3.7 (2.4-6.2)μM, 9.8 (7.9-12.2)μM, and 41.1 (32.5-52.1)μM, respectively. Modest but statistically significant correlations were noted between TMAO and choline (r = 0.33, P < 0.001) and less between TMAO and betaine (r = 0.09, P < 0.001). Higher plasma choline and betaine levels were associated with a 1.9-fold and 1.4-fold increased risk of MACE, respectively (Quartiles 4 vs. 1; P < 0.01, each). Following adjustments for traditional cardiovascular risk factors and high-sensitivity C-reactive protein, elevated choline [1.34 (1.03-1.74), P < 0.05], and betaine levels [1.33 (1.03-1.73), P < 0.05] each predicted increased MACE risk. Neither choline nor betaine predicted MACE risk when TMAO was added to the adjustment model, and choline and betaine predicted future risk for MACE only when TMAO was elevated.Elevated plasma levels of choline and betaine are each associated with incident MACE risk independent of traditional risk factors. However, high choline and betaine levels are only associated with higher risk of future MACE with concomitant increase in TMAO.
DOI: 10.1056/nejmoa1510774
2015
Cited 452 times
Isosorbide Mononitrate in Heart Failure with Preserved Ejection Fraction
Nitrates are commonly prescribed to enhance activity tolerance in patients with heart failure and a preserved ejection fraction. We compared the effect of isosorbide mononitrate or placebo on daily activity in such patients.In this multicenter, double-blind, crossover study, 110 patients with heart failure and a preserved ejection fraction were randomly assigned to a 6-week dose-escalation regimen of isosorbide mononitrate (from 30 mg to 60 mg to 120 mg once daily) or placebo, with subsequent crossover to the other group for 6 weeks. The primary end point was the daily activity level, quantified as the average daily accelerometer units during the 120-mg phase, as assessed by patient-worn accelerometers. Secondary end points included hours of activity per day during the 120-mg phase, daily accelerometer units during all three dose regimens, quality-of-life scores, 6-minute walk distance, and levels of N-terminal pro-brain natriuretic peptide (NT-proBNP).In the group receiving the 120-mg dose of isosorbide mononitrate, as compared with the placebo group, there was a nonsignificant trend toward lower daily activity (-381 accelerometer units; 95% confidence interval [CI], -780 to 17; P=0.06) and a significant decrease in hours of activity per day (-0.30 hours; 95% CI, -0.55 to -0.05; P=0.02). During all dose regimens, activity in the isosorbide mononitrate group was lower than that in the placebo group (-439 accelerometer units; 95% CI, -792 to -86; P=0.02). Activity levels decreased progressively and significantly with increased doses of isosorbide mononitrate (but not placebo). There were no significant between-group differences in the 6-minute walk distance, quality-of-life scores, or NT-proBNP levels.Patients with heart failure and a preserved ejection fraction who received isosorbide mononitrate were less active and did not have better quality of life or submaximal exercise capacity than did patients who received placebo. (Funded by the National Heart, Lung, and Blood Institute; ClinicalTrials.gov number, NCT02053493.).
DOI: 10.1038/s41569-018-0108-7
2018
Cited 446 times
Dietary metabolism, the gut microbiome, and heart failure
Advances in our understanding of how the gut microbiota contributes to human health and diseases have expanded our insight into how microbial composition and function affect the human host. Heart failure is associated with splanchnic circulation congestion, leading to bowel wall oedema and impaired intestinal barrier function. This situation is thought to heighten the overall inflammatory state via increased bacterial translocation and the presence of bacterial products in the systemic blood circulation. Several metabolites produced by gut microorganisms from dietary metabolism have been linked to pathologies such as atherosclerosis, hypertension, heart failure, chronic kidney disease, obesity, and type 2 diabetes mellitus. These findings suggest that the gut microbiome functions like an endocrine organ by generating bioactive metabolites that can directly or indirectly affect host physiology. In this Review, we discuss several newly discovered gut microbial metabolic pathways, including the production of trimethylamine and trimethylamine N-oxide, short-chain fatty acids, and secondary bile acids, that seem to participate in the development and progression of cardiovascular diseases, including heart failure. We also discuss the gut microbiome as a novel therapeutic target for the treatment of cardiovascular disease, and potential strategies for targeting intestinal microbial processes. Metabolites produced by gut microorganisms from dietary metabolism have been linked to the pathogenesis of heart failure, suggesting that the gut microbiome functions like an endocrine organ. In this Review, Tang and colleagues discuss the gut microbial metabolic pathways involved in heart failure and propose the gut microbiome as a therapeutic target.
DOI: 10.1016/j.amjmed.2008.03.046
2008
Cited 438 times
Diabetic Cardiomyopathy: Insights into Pathogenesis, Diagnostic Challenges, and Therapeutic Options
Diabetic cardiomyopathy is the presence of myocardial dysfunction in the absence of coronary artery disease and hypertension. Hyperglycemia seems to be central to the pathogenesis of diabetic cardiomyopathy and to trigger a series of maladaptive stimuli that result in myocardial fibrosis and collagen deposition. These processes are thought to be responsible for altered myocardial relaxation characteristics and manifest as diastolic dysfunction on imaging. Sophisticated imaging technologies also have permitted the detection of subtle systolic dysfunction in the diabetic myocardium. In the early stages, these changes appear reversible with tight metabolic control, but as the pathologic processes become organized, the changes are irreversible and contribute to an excess risk of heart failure among diabetic patients independently of common comorbidities, such as coronary artery disease and hypertension. Therapeutic agents specifically targeting processes that lead to these pathophysiologic changes are in the early stages of development. Although glycemic control and early administration of neurohormonal antagonists remain the cornerstones of therapeutic approaches, newer treatment targets are currently being explored.
DOI: 10.1016/j.cmet.2014.10.006
2014
Cited 423 times
γ-Butyrobetaine Is a Proatherogenic Intermediate in Gut Microbial Metabolism of L-Carnitine to TMAO
L-carnitine, a nutrient in red meat, was recently reported to accelerate atherosclerosis via a metaorganismal pathway involving gut microbial trimethylamine (TMA) formation and host hepatic conversion into trimethylamine-N-oxide (TMAO). Herein, we show that following L-carnitine ingestion, γ-butyrobetaine (γBB) is produced as an intermediary metabolite by gut microbes at a site anatomically proximal to and at a rate ∼1,000-fold higher than the formation of TMA. Moreover, we show that γBB is the major gut microbial metabolite formed from dietary L-carnitine in mice, is converted into TMA and TMAO in a gut microbiota-dependent manner (like dietary L-carnitine), and accelerates atherosclerosis. Gut microbial composition and functional metabolic studies reveal that distinct taxa are associated with the production of γBB or TMA/TMAO from dietary L-carnitine. Moreover, despite their close structural similarity, chronic dietary exposure to L-carnitine or γBB promotes development of functionally distinct microbial communities optimized for the metabolism of L-carnitine or γBB, respectively.
DOI: 10.1016/j.jacc.2008.11.024
2009
Cited 421 times
Insights From a Cardiac Resynchronization Optimization Clinic as Part of a Heart Failure Disease Management Program
Our aim was to determine the feasibility and value of a protocol-driven approach to patients with cardiac resynchronization therapy (CRT) who did not exhibit a positive response long after implant.Up to one-third of patients with advanced heart failure do not exhibit a positive response to CRT.A total of 75 consecutive ambulatory patients with persistent advanced heart failure symptoms and/or adverse reverse remodeling and CRT implanted >6 months underwent a comprehensive protocol-driven evaluation to determine the potential reasons for a suboptimal response. Recommendations were made to maximize the potential of CRT, and adverse events were documented.All patients (mean left ventricular [LV] ejection fraction 23 +/- 9%, LV end-diastolic volume 275 +/- 127 ml) underwent evaluation. Eighty-eight percent of patients had significantly better echocardiographic indexes of LV filling and LV ejection with optimal setting of their CRT compared with a temporary VVI back-up setting. Most patients had identifiable reasons for suboptimal response, including inadequate device settings (47%), suboptimal medical treatment (32%), arrhythmias (32%), inappropriate lead position (21%), or lack of baseline dyssynchrony (9%). Multidisciplinary recommendations led to changes in device settings and/or other therapy modifications in 74% of patients and were associated with fewer adverse events (13% vs. 50%, odds ratio: 0.2 [95% confidence interval: 0.07 to 0.56], p = 0.002) compared with those in which no recommendation could be made.Routine protocol-driven approach to evaluate ambulatory CRT patients who did not exhibit a positive response is feasible, and changes in device settings and/or other therapies after multidisciplinary evaluation may be associated with fewer adverse events.
DOI: 10.1001/jama.2013.282190
2013
Cited 418 times
Low-Dose Dopamine or Low-Dose Nesiritide in Acute Heart Failure With Renal Dysfunction
<h3>Importance</h3> Small studies suggest that low-dose dopamine or low-dose nesiritide may enhance decongestion and preserve renal function in patients with acute heart failure and renal dysfunction; however, neither strategy has been rigorously tested. <h3>Objective</h3> To test the 2 independent hypotheses that, compared with placebo, addition of low-dose dopamine (2 μg/kg/min) or low-dose nesiritide (0.005 μg/kg/min without bolus) to diuretic therapy will enhance decongestion and preserve renal function in patients with acute heart failure and renal dysfunction. <h3>Design, Setting, and Participants</h3> Multicenter, double-blind, placebo-controlled clinical trial (Renal Optimization Strategies Evaluation [ROSE]) of 360 hospitalized patients with acute heart failure and renal dysfunction (estimated glomerular filtration rate of 15-60 mL/min/1.73 m<sup>2</sup>), randomized within 24 hours of admission. Enrollment occurred from September 2010 to March 2013 across 26 sites in North America. <h3>Interventions</h3> Participants were randomized in an open, 1:1 allocation ratio to the dopamine or nesiritide strategy. Within each strategy, participants were randomized in a double-blind, 2:1 ratio to active treatment or placebo. The dopamine (n = 122) and nesiritide (n = 119) groups were independently compared with the pooled placebo group (n = 119). <h3>Main Outcomes and Measures</h3> Coprimary end points included 72-hour cumulative urine volume (decongestion end point) and the change in serum cystatin C from enrollment to 72 hours (renal function end point). <h3>Results</h3> Compared with placebo, low-dose dopamine had no significant effect on 72-hour cumulative urine volume (dopamine, 8524 mL; 95% CI, 7917-9131 vs placebo, 8296 mL; 95% CI, 7762-8830 ; difference, 229 mL; 95% CI, −714 to 1171 mL;<i>P</i> = .59) or on the change in cystatin C level (dopamine, 0.12 mg/L; 95% CI, 0.06-0.18 vs placebo, 0.11 mg/L; 95% CI, 0.06-0.16; difference, 0.01; 95% CI, −0.08 to 0.10;<i>P</i> = .72). Similarly, low-dose nesiritide had no significant effect on 72-hour cumulative urine volume (nesiritide, 8574 mL; 95% CI, 8014-9134 vs placebo, 8296mL; 95% CI, 7762-8830; difference, 279 mL; 95% CI, −618 to 1176 mL;<i>P</i> = .49) or on the change in cystatin C level (nesiritide, 0.07 mg/L; 95% CI, 0.01-0.13 vs placebo, 0.11 mg/L; 95% CI, 0.06-0.16; difference, −0.04; 95% CI, −0.13 to 0.05;<i>P</i> = .36). Compared with placebo, there was no effect of low-dose dopamine or nesiritide on secondary end points reflective of decongestion, renal function, or clinical outcomes. <h3>Conclusion and Relevance</h3> In participants with acute heart failure and renal dysfunction, neither low-dose dopamine nor low-dose nesiritide enhanced decongestion or improved renal function when added to diuretic therapy. <h3>Trial Registration</h3> clinicaltrials.gov Identifier:NCT01132846
DOI: 10.1016/j.cell.2020.02.016
2020
Cited 399 times
A Cardiovascular Disease-Linked Gut Microbial Metabolite Acts via Adrenergic Receptors
Using untargeted metabolomics (n = 1,162 subjects), the plasma metabolite (m/z = 265.1188) phenylacetylglutamine (PAGln) was discovered and then shown in an independent cohort (n = 4,000 subjects) to be associated with cardiovascular disease (CVD) and incident major adverse cardiovascular events (myocardial infarction, stroke, or death). A gut microbiota-derived metabolite, PAGln, was shown to enhance platelet activation-related phenotypes and thrombosis potential in whole blood, isolated platelets, and animal models of arterial injury. Functional and genetic engineering studies with human commensals, coupled with microbial colonization of germ-free mice, showed the microbial porA gene facilitates dietary phenylalanine conversion into phenylacetic acid, with subsequent host generation of PAGln and phenylacetylglycine (PAGly) fostering platelet responsiveness and thrombosis potential. Both gain- and loss-of-function studies employing genetic and pharmacological tools reveal PAGln mediates cellular events through G-protein coupled receptors, including α2A, α2B, and β2-adrenergic receptors. PAGln thus represents a new CVD-promoting gut microbiota-dependent metabolite that signals via adrenergic receptors.Video AbstracteyJraWQiOiI4ZjUxYWNhY2IzYjhiNjNlNzFlYmIzYWFmYTU5NmZmYyIsImFsZyI6IlJTMjU2In0.eyJzdWIiOiI1Y2Q0ZGZhMTNjOWZjODc1ZmQwODJkN2ZhMGZjY2M4OSIsImtpZCI6IjhmNTFhY2FjYjNiOGI2M2U3MWViYjNhYWZhNTk2ZmZjIiwiZXhwIjoxNjc4NTMyOTIxfQ.A7ok2qU77Vr0vgq8j1GPEn0w0uDprE84fuTG3Odr3YHzobK5z0XznViS5PD96VB3Dg1F02Gv-jBxQH-IXc8fryspRQcWNm2KBxypipWF8UeKgaKtAZfye0cRomFeYI5_Iza4mBs9260GDGX112uiizfXMLf_IQV742wepwhstcvKhiq9f2ovi9mQArvBrCO1TWP_-dAEEJO2FElitNSRqkmvZoRQJHwiANR-XGDBdqjQ3ogtfKipDUdkSYz0-BZT29l1rCYWxFY_0S1QDnxc0CZVjuhacEiFOWfmCVZVBIETFqZFE3xtqoqmPQqW80ki4DBLK5KB6xG7plHLvP9i2w(mp4, (36.03 MB) Download video
DOI: 10.1161/circulationaha.108.779223
2009
Cited 397 times
Tissue Doppler Imaging in the Estimation of Intracardiac Filling Pressure in Decompensated Patients With Advanced Systolic Heart Failure
The ratio of early transmitral velocity to tissue Doppler mitral annular early diastolic velocity (E/Ea) has been correlated with pulmonary capillary wedge pressure (PCWP) in a wide variety of cardiac conditions. The objective of this study was to determine the reliability of mitral E/Ea for predicting PCWP in patients admitted for advanced decompensated heart failure.Prospective consecutive patients with advanced decompensated heart failure (ejection fraction < or =30%, New York Heart Association class III to IV symptoms) underwent simultaneous echocardiographic and hemodynamic evaluation on admission and after 48 hours of intensive medical therapy. A total of 106 patients were included (mean age, 57+/-12 years; ejection fraction, 24+/-8%; PCWP, 21+/-7 mm Hg; mitral E/Ea ratio, 20+/-12). No correlation was found between mitral E/Ea ratio and PCWP, particularly in those with larger left ventricular volumes, more impaired cardiac indexes, and the presence of cardiac resynchronization therapy. Overall, the mitral E/Ea ratio was similar among patients with PCWP >18 and < or =18 mm Hg, and sensitivity and specificity for mitral E/Ea ratio >15 to identify a PCWP >18 mm Hg were 66% and 50%, respectively. Contrary to prior reports, we did not observe any direct association between changes in PCWP and changes in mitral E/Ea ratio.In decompensated patients with advanced systolic heart failure, tissue Doppler-derived mitral E/Ea ratio may not be as reliable in predicting intracardiac filling pressures, particularly in those with larger LV volumes, more impaired cardiac indices, and the presence of cardiac resynchronization therapy.
DOI: 10.1016/s0735-1097(02)02608-6
2002
Cited 388 times
Cardiac troponins in renal insufficiency
Patients with renal insufficiency may have increased serum troponins even in the absence of clinically suspected acute myocardial ischemia. While cardiovascular disease is the most common cause of death in patients with renal failure, we are just beginning to understand the clinical meaning of serum troponin elevations. Serum troponin T is increased more frequently than troponin I in patients with renal failure, leading clinicians to question its specificity for the diagnosis of myocardial infarction. Many large-scale trials demonstrating the utility of serum troponins in predicting adverse events and in guiding therapy and intervention in acute coronary syndromes have excluded patients with renal failure. Despite persistent uncertainty about the mechanism of elevated serum troponins in patients with reduced renal function, data from smaller groups of renal failure patients have suggested that troponin elevations are associated with added risk, including an increase in mortality. It is possible that increases in serum troponin from baseline in patients with renal insufficiency admitted to hospital with acute coronary syndrome may signify myocardial necrosis. Further studies are needed to clarify this hypothesis.
DOI: 10.1016/j.jacc.2007.09.043
2008
Cited 377 times
Elevated Intra-Abdominal Pressure in Acute Decompensated Heart Failure
This study sought to determine whether changes in intra-abdominal pressure (IAP) with aggressive diuretic or vasodilator therapy are associated with improvement in renal function in acute decompensated heart failure (ADHF). Elevated IAP (≥8 mm Hg) is associated with intra-abdominal organ dysfunction. There is potential for ascites and visceral edema causing elevated IAP in patients with ADHF. Forty consecutive patients admitted to a specialized heart failure intensive care unit for management of ADHF with intensive medical therapy were studied. The IAP was measured using a simple transvesical technique at time of admission and before removal of the pulmonary artery catheter. In our study cohort (mean age 59 ± 13 years, mean left ventricular ejection fraction 19 ± 9%, baseline serum creatinine 2.0 ± 0.9 mg/dl), the mean baseline IAP was 8 ± 4 mm Hg, with 24 (60%) patients having elevated IAP. Elevated IAP was associated with worse renal function (p = 0.009). Intensive medical therapy resulted in improvement in both hemodynamic measurements and IAP. A strong correlation (r = 0.77, p < 0.001) was observed between reduction in IAP and improved renal function in patients with baseline elevated IAP. However, changes in IAP or renal function did not correlate with changes in any hemodynamic variable. Elevated IAP is prevalent in patients with ADHF and is associated with impaired renal function. In the setting of intensive medical therapy for ADHF, changes in IAP were better correlated with changes in renal function than any hemodynamic variable.
DOI: 10.1093/jnci/djq091
2010
Cited 374 times
Initial Assessment, Surveillance, and Management of Blood Pressure in Patients Receiving Vascular Endothelial Growth Factor Signaling Pathway Inhibitors
Hypertension is a mechanism-based toxic effect of drugs that inhibit the vascular endothelial growth factor signaling pathway (VSP). Substantial evidence exists for managing hypertension as a chronic condition, but there are few prospectively collected data on managing acute hypertension caused by VSP inhibitors. The Investigational Drug Steering Committee of the National Cancer Institute convened an interdisciplinary cardiovascular toxicities expert panel to evaluate this problem, to make recommendations to the Cancer Therapy Evaluation Program on further study, and to structure an approach for safe management by treating physicians. The panel reviewed: the published literature on blood pressure (BP), hypertension, and specific VSP inhibitors; abstracts from major meetings; shared experience with the development of VSP inhibitors; and established principles of hypertension care. The panel generated a consensus report including the recommendations on clinical concerns summarized here. To support the greatest possible number of patients to receive VSP inhibitors safely and effectively, the panel had four recommendations: 1) conduct and document a formal risk assessment for potential cardiovascular complications, 2) recognize that preexisting hypertension will be common in cancer patients and should be identified and addressed before initiation of VSP inhibitor therapy, 3) actively monitor BP throughout treatment with more frequent assessments during the first cycle of treatment, and 4) manage BP with a goal of less than 140/90 mmHg for most patients (and to lower, prespecified goals in patients with specific preexisting cardiovascular risk factors). Proper agent selection, dosing, and scheduling of follow-up should enable maintaining VSP inhibition while avoiding the complications associated with excessive or prolonged elevation in BP.
DOI: 10.1373/clinchem.2006.084194
2007
Cited 363 times
National Academy of Clinical Biochemistry Laboratory Medicine Practice Guidelines: Clinical Characteristics and Utilization of Biochemical Markers in Acute Coronary Syndromes
### a. definition of terms Acute coronary syndrome (ACS)1 refers to a constellation of clinical symptoms caused by acute myocardial ischemia (1)(2). Owing to their higher risk for cardiac death or ischemic complications, patients with ACS must be identified among the estimated 8 million patients with nontraumatic chest symptoms presenting for emergency evaluation each year in the US (3). In practice, the terms suspected or possible ACS are often used by medical personnel early in the process of evaluation to describe patients for whom the symptom complex is consistent with ACS but the diagnosis has not yet been conclusively established (1). Patients with ACS are subdivided into 2 major categories based on the 12-lead electrocardiogram (ECG) at presentation (Fig. 1⇓ ): those with new ST-segment …
DOI: 10.1001/jama.2017.5427
2017
Cited 334 times
Effect of Oral Iron Repletion on Exercise Capacity in Patients With Heart Failure With Reduced Ejection Fraction and Iron Deficiency
<h3>Importance</h3> Iron deficiency is present in approximately 50% of patients with heart failure with reduced left ventricular ejection fraction (HFrEF) and is an independent predictor of reduced functional capacity and mortality. However, the efficacy of inexpensive readily available oral iron supplementation in heart failure is unknown. <h3>Objective</h3> To test whether therapy with oral iron improves peak exercise capacity in patients with HFrEF and iron deficiency. <h3>Design, Setting, and Participants</h3> Phase 2, double-blind, placebo-controlled randomized clinical trial of patients with HFrEF (&lt;40%) and iron deficiency, defined as a serum ferritin level of 15 to 100 ng/mL or a serum ferritin level of 101 to 299 ng/mL with transferrin saturation of less than 20%. Participants were enrolled between September 2014 and November 2015 at 23 US sites. <h3>Interventions</h3> Oral iron polysaccharide (n = 111) or placebo (n = 114), 150 mg twice daily for 16 weeks. <h3>Main Outcomes and Measures</h3> The primary end point was a change in peak oxygen uptake (V̇o<sub>2</sub>) from baseline to 16 weeks. Secondary end points were change in 6-minute walk distance, plasma N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels, and health status as assessed by Kansas City Cardiomyopathy Questionnaire (KCCQ, range 0-100, higher scores reflect better quality of life). <h3>Results</h3> Among 225 randomized participants (median age, 63 years; 36% women) 203 completed the study. The median baseline peak V̇o<sub>2</sub>was 1196 mL/min (interquartile range [IQR], 887-1448 mL/min) in the oral iron group and 1167 mL/min (IQR, 887-1449 mL/min) in the placebo group. The primary end point, change in peak V̇o<sub>2</sub>at 16 weeks, did not significantly differ between the oral iron and placebo groups (+23 mL/min vs −2 mL/min; difference, 21 mL/min [95% CI, −34 to +76 mL/min];<i>P</i> = .46). Similarly, at 16 weeks, there were no significant differences between treatment groups in changes in 6-minute walk distance (−13 m; 95% CI, −32 to 6 m), NT-proBNP levels (159; 95% CI, −280 to 599 pg/mL), or KCCQ score (1; 95% CI, −2.4 to 4.4), all<i>P</i> &gt; .05. <h3>Conclusions and Relevance</h3> Among participants with HFrEF with iron deficiency, high-dose oral iron did not improve exercise capacity over 16 weeks. These results do not support use of oral iron supplementation in patients with HFrEF. <h3>Trial Registration</h3> clinicaltrials.govIdentifier:NCT02188784
DOI: 10.1161/circheartfailure.110.958223
2011
Cited 329 times
High-Sensitivity ST2 for Prediction of Adverse Outcomes in Chronic Heart Failure
Background— Soluble ST2 reflects activity of an interleukin-33–dependent cardioprotective signaling axis and is a diagnostic and prognostic marker in acute heart failure. The use of ST2 in chronic heart failure has not been well defined. Our objective was to determine whether plasma ST2 levels predict adverse outcomes in chronic heart failure in the context of current approaches. Methods and Results— We determined the association between ST2 level and risk of death or transplantation in a multicenter, prospective cohort of 1141 chronic heart failure outpatients. Adjusted Cox models, receiver operating characteristic analyses, and risk reclassification metrics were used to assess the value of ST2 in predicting risk beyond currently used factors. After a median of 2.8 years, 267 patients (23%) died or underwent heart transplantation. Patients in the highest ST2 tertile (ST2 &gt;36.3 ng/mL) had a markedly increased risk of adverse outcomes compared with the lowest tertile (ST2 ≤22.3 ng/mL), with an unadjusted hazard ratio of 3.2 (95% confidence interval [CI], 2.2 to 4.7; P &lt;0.0001) that remained significant after multivariable adjustment (adjusted hazard ratio, 1.9; 95% CI, 1.3 to 2.9; P =0.002). In receiver operating characteristic analyses, the area under the curve for ST2 was 0.75 (95% CI, 0.69 to 0.79), which was similar to N-terminal pro-B–type natriuretic peptide (NT-proBNP) (area under the curve, 0.77; 95% CI, 0.72 to 0.81; P =0.24 versus ST2) but lower than the Seattle Heart Failure Model (area under the curve, 0.81 (95% CI, 0.77 to 0.85; P =0.014 versus ST2). Addition of ST2 and NT-proBNP to the Seattle Heart Failure Model reclassified 14.9% of patients into more appropriate risk categories ( P =0.017). Conclusions— ST2 is a potent marker of risk in chronic heart failure and when used in combination with NT-proBNP offers moderate improvement in assessing prognosis beyond clinical risk scores.
DOI: 10.1038/nm.3459
2014
Cited 319 times
An abundant dysfunctional apolipoprotein A1 in human atheroma
Recent studies have indicated that high-density lipoproteins (HDLs) and their major structural protein, apolipoprotein A1 (apoA1), recovered from human atheroma are dysfunctional and are extensively oxidized by myeloperoxidase (MPO). In vitro oxidation of either apoA1 or HDL particles by MPO impairs their cholesterol acceptor function. Here, using phage display affinity maturation, we developed a high-affinity monoclonal antibody that specifically recognizes both apoA1 and HDL that have been modified by the MPO-H2O2-Cl(-) system. An oxindolyl alanine (2-OH-Trp) moiety at Trp72 of apoA1 is the immunogenic epitope. Mutagenesis studies confirmed a critical role for apoA1 Trp72 in MPO-mediated inhibition of the ATP-binding cassette transporter A1 (ABCA1)-dependent cholesterol acceptor activity of apoA1 in vitro and in vivo. ApoA1 containing a 2-OH-Trp72 group (oxTrp72-apoA1) is in low abundance within the circulation but accounts for 20% of the apoA1 in atherosclerosis-laden arteries. OxTrp72-apoA1 recovered from human atheroma or plasma is lipid poor, virtually devoid of cholesterol acceptor activity and demonstrated both a potent proinflammatory activity on endothelial cells and an impaired HDL biogenesis activity in vivo. Elevated oxTrp72-apoA1 levels in subjects presenting to a cardiology clinic (n = 627) were associated with increased cardiovascular disease risk. Circulating oxTrp72-apoA1 levels may serve as a way to monitor a proatherogenic process in the artery wall.
DOI: 10.1016/j.jacc.2013.04.070
2013
Cited 309 times
Abdominal Contributions to Cardiorenal Dysfunction in Congestive Heart Failure
Current pathophysiological models of congestive heart failure unsatisfactorily explain the detrimental link between congestion and cardiorenal function. Abdominal congestion (i.e., splanchnic venous and interstitial congestion) manifests in a substantial number of patients with advanced congestive heart failure, yet is poorly defined. Compromised capacitance function of the splanchnic vasculature and deficient abdominal lymph flow resulting in interstitial edema might both be implied in the occurrence of increased cardiac filling pressures and renal dysfunction. Indeed, increased intra-abdominal pressure, as an extreme marker of abdominal congestion, is correlated with renal dysfunction in advanced congestive heart failure. Intriguing findings provide preliminary evidence that alterations in the liver and spleen contribute to systemic congestion in heart failure. Finally, gut-derived hormones might influence sodium homeostasis, whereas entrance of bowel toxins into the circulatory system, as a result of impaired intestinal barrier function secondary to congestion, might further depress cardiac as well as renal function. Those toxins are mainly produced by micro-organisms in the gut lumen, with presumably important alterations in advanced heart failure, especially when renal function is depressed. Therefore, in this state-of-the-art review, we explore the crosstalk between the abdomen, heart, and kidneys in congestive heart failure. This might offer new diagnostic opportunities as well as treatment strategies to achieve decongestion in heart failure, especially when abdominal congestion is present. Among those currently under investigation are paracentesis, ultrafiltration, peritoneal dialysis, oral sodium binders, vasodilator therapy, renal sympathetic denervation and agents targeting the gut microbiota.
DOI: 10.1093/eurheartj/ehy799
2018
Cited 301 times
Impact of chronic dietary red meat, white meat, or non-meat protein on trimethylamine N-oxide metabolism and renal excretion in healthy men and women
AimsCarnitine and choline are major nutrient precursors for gut microbiota-dependent generation of the atherogenic metabolite, trimethylamine N-oxide (TMAO). We performed randomized-controlled dietary intervention studies to explore the impact of chronic dietary patterns on TMAO levels, metabolism and renal excretion.
DOI: 10.1016/j.jacc.2019.03.024
2019
Cited 300 times
Intestinal Microbiota in Cardiovascular Health and Disease
Despite major strides in reducing cardiovascular disease (CVD) burden with modification of classic CVD risk factors, significant residual risks remain. Recent discoveries that linked intestinal microbiota and CVD have broadened our understanding of how dietary nutrients may affect cardiovascular health and disease. Although next-generation sequencing techniques can identify gut microbial community participants and provide insights into microbial composition shifts in response to physiological responses and dietary exposures, provisions of prebiotics or probiotics have yet to show therapeutic benefit for CVD. Our evolving understanding of intestinal microbiota-derived physiological modulators (e.g., short-chain fatty acids) and pathogenic mediators (e.g., trimethylamine N-oxide) of host disease susceptibility have created novel potential therapeutic opportunities for improved cardiovascular health. This review discusses the roles of human intestinal microbiota in normal physiology, their associations with CVD susceptibilities, and the potential of modulating intestinal microbiota composition and metabolism as a novel therapeutic target for CVD.
DOI: 10.1371/journal.pgen.1001300
2011
Cited 293 times
Genome-Wide Association Study of Coronary Heart Disease and Its Risk Factors in 8,090 African Americans: The NHLBI CARe Project
Coronary heart disease (CHD) is the leading cause of mortality in African Americans. To identify common genetic polymorphisms associated with CHD and its risk factors (LDL- and HDL-cholesterol (LDL-C and HDL-C), hypertension, smoking, and type-2 diabetes) in individuals of African ancestry, we performed a genome-wide association study (GWAS) in 8,090 African Americans from five population-based cohorts. We replicated 17 loci previously associated with CHD or its risk factors in Caucasians. For five of these regions (CHD: CDKN2A/CDKN2B; HDL-C: FADS1-3, PLTP, LPL, and ABCA1), we could leverage the distinct linkage disequilibrium (LD) patterns in African Americans to identify DNA polymorphisms more strongly associated with the phenotypes than the previously reported index SNPs found in Caucasian populations. We also developed a new approach for association testing in admixed populations that uses allelic and local ancestry variation. Using this method, we discovered several loci that would have been missed using the basic allelic and global ancestry information only. Our conclusions suggest that no major loci uniquely explain the high prevalence of CHD in African Americans. Our project has developed resources and methods that address both admixture- and SNP-association to maximize power for genetic discovery in even larger African-American consortia.
DOI: 10.1161/atvbaha.113.301373
2013
Cited 273 times
Paradoxical Association of Enhanced Cholesterol Efflux With Increased Incident Cardiovascular Risks
Objective— Diminished cholesterol efflux activity of apolipoprotein B (apoB)–depleted serum is associated with prevalent coronary artery disease, but its prognostic value for incident cardiovascular events is unclear. We investigated the relationship of cholesterol efflux activity with both prevalent coronary artery disease and incident development of major adverse cardiovascular events (death, myocardial infarction, or stroke). Approach and Results— Cholesterol efflux activity from free cholesterol–enriched macrophages was measured in 2 case–control cohorts: (1) an angiographic cohort (n=1150) comprising stable subjects undergoing elective diagnostic coronary angiography and (2) an outpatient cohort (n=577). Analysis of media from cholesterol efflux assays revealed that the high-density lipoprotein fraction (1.063&lt; d &lt;1.21) contained only a minority (≈40%) of [ 14 C]cholesterol released, with the majority found within the lipoprotein particle–depleted fraction, where ≈60% was recovered after apolipoprotein A1 immunoprecipitation. Albumin immunoprecipitation recovered another ≈30% of radiolabeled cholesterol within this fraction. Enhanced cholesterol efflux activity from ATP-binding cassette transporter A1–stimulated macrophages was associated with reduced risk of prevalent coronary artery disease in unadjusted models within both cohorts; however, the inverse risk relationship remained significant after adjustment for traditional coronary artery disease risk factors only within the outpatient cohort. Surprisingly, higher cholesterol efflux activity was associated with increase in prospective (3 years) risk of myocardial infarction/stroke (adjusted hazard ratio, 2.19; 95% confidence interval, 1.02–4.74) and major adverse cardiovascular events (adjusted hazard ratio, 1.85; 95% confidence interval, 1.11–3.06). Conclusions— Heightened cholesterol efflux to apoB-depleted serum was paradoxically associated with increased prospective risk for myocardial infarction, stroke, and death. The majority of released radiolabeled cholesterol from macrophages in cholesterol efflux activity assays does not reside within a high-density lipoprotein particle.
DOI: 10.1016/j.cardfail.2014.11.006
2015
Cited 268 times
Intestinal Microbiota-Dependent Phosphatidylcholine Metabolites, Diastolic Dysfunction, and Adverse Clinical Outcomes in Chronic Systolic Heart Failure
Trimethylamine-N-oxide (TMAO) has been linked to increased cardiovascular risk. We aimed to determine the prognostic value of TMAO and its dietary precursors, choline and betaine, in heart failure (HF).In 112 patients with chronic systolic HF with comprehensive echocardiographic evaluation, we measured plasma TMAO, choline, and betaine by mass spectrometry. Median (interquartile range) TMAO levels, choline, and betaine levels were 5.8 (3.6-12.1) μmol/L, 10.9 (8.4-14.0) μmol/L, and 43.8 (37.1-53.0) μmol/L, respectively, and were correlated with each other (all P < .0001 for both). TMAO levels were significantly higher in patients with diabetes mellitus (9.4 [4.9-13.2] vs 4.8 [3.4-9.8] μmol/L; P = .005) and in subjects with New York Heart Association functional class III or greater (7.0 [4.7-14.8] vs 4.7 [3.4-11.3] μmol/L; P = .02). Elevated TMAO, choline, and betaine levels were each associated with higher plasma N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels and more advanced left ventricular diastolic dysfunction, but not systolic dysfunction or inflammatory and endothelial biomarkers. Higher choline (hazard ratio [HR] 1.64, 95% CI 1.22-2.20; P = .001), betaine (HR 1.51, 95% CI 1.10-2.08; P = .01), and TMAO (HR 1.48, 95% CI 1.10-1.96; P = .01) predicted increased risk for 5-year adverse clinical events (death/transplantation). Only higher TMAO levels predicted incident adverse clinical events independently from age, estimated glomerular filtration rate, mitral E/septal Ea, and NT-proBNP levels (HR 1.46, 95% CI 1.03-2.14; P = .03).Elevated plasma TMAO, choline, and betaine levels are each associated with more advanced left ventricular diastolic dysfunction and portend poorer long-term adverse clinical outcomes in chronic systolic HF. However, only higher plasma TMAO was associated with poor prognosis after adjustment for cardiorenal indices.
DOI: 10.1161/circheartfailure.115.002314
2016
Cited 263 times
Choline Diet and Its Gut Microbe–Derived Metabolite, Trimethylamine N-Oxide, Exacerbate Pressure Overload–Induced Heart Failure
Trimethylamine N-oxide (TMAO), a gut microbe-dependent metabolite of dietary choline and other trimethylamine-containing nutrients, is both elevated in the circulation of patients having heart failure and heralds worse overall prognosis. In animal studies, dietary choline or TMAO significantly accelerates atherosclerotic lesion development in ApoE-deficient mice, and reduction in TMAO levels inhibits atherosclerosis development in the low-density lipoprotein receptor knockout mouse.C57BL6/J mice were fed either a control diet, a diet containing choline (1.2%) or a diet containing TMAO (0.12%) starting 3 weeks before surgical transverse aortic constriction. Mice were studied for 12 weeks after transverse aortic constriction. Cardiac function and left ventricular structure were monitored at 3-week intervals using echocardiography. Twelve weeks post transverse aortic constriction, myocardial tissues were collected to evaluate cardiac and vascular fibrosis, and blood samples were evaluated for cardiac brain natriuretic peptide, choline, and TMAO levels. Pulmonary edema, cardiac enlargement, and left ventricular ejection fraction were significantly (P<0.05, each) worse in mice fed either TMAO- or choline-supplemented diets when compared with the control diet. In addition, myocardial fibrosis was also significantly greater (P<0.01, each) in the TMAO and choline groups relative to controls.Heart failure severity is significantly enhanced in mice fed diets supplemented with either choline or the gut microbe-dependent metabolite TMAO. The present results suggest that additional studies are warranted examining whether gut microbiota and the dietary choline → TMAO pathway contribute to increased heart failure susceptibility.
DOI: 10.1016/j.jacc.2013.05.020
2013
Cited 254 times
2013 ACCF/AHA Guideline for the Management of Heart Failure: Executive Summary
DOI: 10.1093/eurheartj/ehw582
2017
Cited 244 times
Gut microbiota-dependent trimethylamine N-oxide in acute coronary syndromes: a prognostic marker for incident cardiovascular events beyond traditional risk factors
Systemic levels of trimethylamine N-oxide (TMAO), a pro-atherogenic and pro-thrombotic metabolite produced from gut microbiota metabolism of dietary trimethylamine (TMA)-containing nutrients such as choline or carnitine, predict incident cardiovascular event risks in stable primary and secondary prevention subjects. However, the prognostic value of TMAO in the setting of acute coronary syndromes (ACS) remains unknown.We investigated the relationship of TMAO levels with incident cardiovascular risks among sequential patients presenting with ACS in two independent cohorts. In the Cleveland Cohort, comprised of sequential subjects (n = 530) presenting to the Emergency Department (ED) with chest pain of suspected cardiac origin, an elevated plasma TMAO level at presentation was independently associated with risk of major adverse cardiac events (MACE, including myocardial infarction, stroke, need for revascularization, or death) over the ensuing 30-day (4th quartile (Q4) adjusted odds ratio (OR) 6.30, 95% confidence interval (CI), 1.89-21.0, P < 0.01) and 6-month (Q4 adjusted OR 5.65, 95%CI, 1.91-16.7; P < 0.01) intervals. TMAO levels were also a significant predictor of the long term (7-year) mortality (Q4 adjusted HR 1.81, 95%CI, 1.04-3.15; P < 0.05). Interestingly, TMAO level at initial presentation predicted risk of incident MACE over the near-term (30 days and 6 months) even among subjects who were initially negative for troponin T (< 0.1 ng/mL) (30 days, Q4 adjusted OR 5.83, 95%CI, 1.79-19.03; P < 0.01). The prognostic value of TMAO was also assessed in an independent multicentre Swiss Cohort of ACS patients (n = 1683) who underwent coronary angiography. Trimethylamine N-oxide again predicted enhanced MACE risk (1-year) (adjusted Q4 hazard ratios: 1.57, 95% CI, 1.03-2.41; P <0.05).Plasma TMAO levels among patients presenting with chest pain predict both near- and long-term risks of incident cardiovascular events, and may thus provide clinical utility in risk stratification among subjects presenting with suspected ACS.
DOI: 10.1161/circulationaha.114.014536
2015
Cited 242 times
Effects of Xanthine Oxidase Inhibition in Hyperuricemic Heart Failure Patients
Oxidative stress may contribute to heart failure (HF) progression. Inhibiting xanthine oxidase in hyperuricemic HF patients may improve outcomes.We randomly assigned 253 patients with symptomatic HF, left ventricular ejection fraction ≤40%, and serum uric acid levels ≥9.5 mg/dL to receive allopurinol (target dose, 600 mg daily) or placebo in a double-blind, multicenter trial. The primary composite end point at 24 weeks was based on survival, worsening HF, and patient global assessment. Secondary end points included change in quality of life, submaximal exercise capacity, and left ventricular ejection fraction. Uric acid levels were significantly reduced with allopurinol in comparison with placebo (treatment difference, -4.2 [-4.9, -3.5] mg/dL and -3.5 [-4.2, -2.7] mg/dL at 12 and 24 weeks, respectively, both P<0.0001). At 24 weeks, there was no significant difference in clinical status between the allopurinol- and placebo-treated patients (worsened 45% versus 46%, unchanged 42% versus 34%, improved 13% versus 19%, respectively; P=0.68). At 12 and 24 weeks, there was no significant difference in change in Kansas City Cardiomyopathy Questionnaire scores or 6-minute walk distances between the 2 groups. At 24 weeks, left ventricular ejection fraction did not change in either group or between groups. Rash occurred more frequently with allopurinol (10% versus 2%, P=0.01), but there was no difference in serious adverse event rates between the groups (20% versus 15%, P=0.36).In high-risk HF patients with reduced ejection fraction and elevated uric acid levels, xanthine oxidase inhibition with allopurinol failed to improve clinical status, exercise capacity, quality of life, or left ventricular ejection fraction at 24 weeks.URL: http://www.clinicaltrials.gov. Unique identifier: NCT00987415.
DOI: 10.1161/circulationaha.117.030112
2018
Cited 238 times
Worsening Renal Function in Patients With Acute Heart Failure Undergoing Aggressive Diuresis Is Not Associated With Tubular Injury
Background: Worsening renal function (WRF) in the setting of aggressive diuresis for acute heart failure treatment may reflect renal tubular injury or simply indicate a hemodynamic or functional change in glomerular filtration. Well-validated tubular injury biomarkers, N -acetyl-β- d -glucosaminidase, neutrophil gelatinase-associated lipocalin, and kidney injury molecule 1, are now available that can quantify the degree of renal tubular injury. The ROSE-AHF trial (Renal Optimization Strategies Evaluation–Acute Heart Failure) provides an experimental platform for the study of mechanisms of WRF during aggressive diuresis for acute heart failure because the ROSE-AHF protocol dictated high-dose loop diuretic therapy in all patients. We sought to determine whether tubular injury biomarkers are associated with WRF in the setting of aggressive diuresis and its association with prognosis. Methods: Patients in the multicenter ROSE-AHF trial with baseline and 72-hour urine tubular injury biomarkers were analyzed (n=283). WRF was defined as a ≥20% decrease in glomerular filtration rate estimated with cystatin C. Results: Consistent with protocol-driven aggressive dosing of loop diuretics, participants received a median 560 mg IV furosemide equivalents (interquartile range, 300–815 mg), which induced a urine output of 8425 mL (interquartile range, 6341–10 528 mL) over the 72-hour intervention period. Levels of N -acetyl-β- d -glucosaminidase and kidney injury molecule 1 did not change with aggressive diuresis (both P &gt;0.59), whereas levels of neutrophil gelatinase-associated lipocalin decreased slightly (−8.7 ng/mg; interquartile range, −169 to 35 ng/mg; P &lt;0.001). WRF occurred in 21.2% of the population and was not associated with an increase in any marker of renal tubular injury: neutrophil gelatinase-associated lipocalin ( P =0.21), N -acetyl-β- d -glucosaminidase ( P =0.46), or kidney injury molecule 1 ( P =0.22). Increases in neutrophil gelatinase-associated lipocalin, N -acetyl-β- d -glucosaminidase, and kidney injury molecule 1 were paradoxically associated with improved survival (adjusted hazard ratio, 0.80 per 10 percentile increase; 95% confidence interval, 0.69–0.91; P =0.001). Conclusions: Kidney tubular injury does not appear to have an association with WRF in the context of aggressive diuresis of patients with acute heart failure. These findings reinforce the notion that the small to moderate deteriorations in renal function commonly encountered with aggressive diuresis are dissimilar from traditional causes of acute kidney injury.
DOI: 10.1016/j.jcmg.2009.08.008
2009
Cited 234 times
Delayed Hyper-Enhancement Magnetic Resonance Imaging Provides Incremental Diagnostic and Prognostic Utility in Suspected Cardiac Amyloidosis
We sought to assess the diagnostic accuracy and incremental prognostic value of delayed hyper-enhancement cardiac magnetic resonance (DHE-CMR) compared with electrocardiographic and transthoracic echocardiographic (TTE) parameters in such patients.Utility of DHE-CMR in the diagnosis of patients with suspected cardiac amyloidosis (CA) has recently been demonstrated, but its incremental prognostic utility is unclear.Forty-seven consecutive patients (mean age 63 years, 70% men, 55% New York Heart Association functional class >II) with suspected CA who underwent electrocardiography (ECG), TTE, DHE-CMR, and biopsy (38 endomyocardial, 9 extracardiac) were studied. Low voltage on ECG was defined as S-wave in lead V(1) + R-wave in lead V(5) or V(6) <15 mm. TTE parameters, including deceleration time, E/E' ratio, and diastolic grade were recorded. CMR was considered positive with diffuse DHE of the subendocardium extending to adjacent myocardium. All-cause mortality was ascertained.In the study population, 59% had low voltage on ECG, 30% had abnormal deceleration time < OR = 150 ms, 38% had E/E' ratio >15, and 47% had advanced (pseudonormal or restrictive) diastology.The diagnostic accuracy of DHE-CMR in patients undergoing endomyocardial biopsy was as follows: sensitivity 88%, specificity 90%, positive predictive value 88%, and negative predictive value 90%. On multivariable logistic regression testing of the diagnostic ability of various noninvasive imaging parameters, only DHE-CMR was significant (Wald chi-square statistic 9.6, p < 0.01). At 1-year post-biopsy, there were 9 (19%) deaths. On Cox proportional hazards analysis, only positive DHE-CMR was a predictor of 1-year mortality (Wald chi-square statistic 4.91, p = 0.03).A characteristic DHE-CMR pattern is more accurate for diagnosis and is a stronger predictor of 1-year mortality in patients with suspected CA as compared with other noninvasive parameters.
DOI: 10.1016/j.jacc.2008.02.088
2008
Cited 234 times
Detection of Soluble Angiotensin-Converting Enzyme 2 in Heart Failure
We sought to determine whether circulating soluble angiotensin-converting enzyme 2 (sACE2) is increased in the plasma of patients with heart failure (HF).Angiotensin-converting enzyme 2 (ACE2) is an integral membrane protein that antagonizes the actions of angiotensin II and prevents the development of HF in animal models. However, because of the need for invasive cardiac tissue sampling, little is known about whether ACE2 is involved in the pathophysiology of HF in humans.We developed a sensitive and specific assay to measure sACE2 activity in human plasma and screened a heterogeneous group of patients suspected of having clinical HF.Increasing sACE2 plasma activity strongly correlated with a clinical diagnosis of HF (p = 0.0002), worsening left ventricular ejection fraction (p < 0.0001), and increasing B-type natriuretic peptide levels (p < 0.0001). Similar to B-type natriuretic peptide, sACE2 activity reflected the severity of HF, with increasing levels associated with worsening New York Heart Association functional class (p < 0.0001). These associations were independent of other disease states and medication use. We found that sACE2 activity was increased in patients with both ischemic and nonischemic cardiomyopathies and also in patients with clinical HF but a preserved left ventricular ejection fraction.Soluble ACE2 activity is increased in patients with HF and correlates with disease severity, suggesting that a cardioprotective arm of the renin-angiotensin-aldosterone system is active in HF.
DOI: 10.1172/jci67478
2013
Cited 228 times
Myeloperoxidase, paraoxonase-1, and HDL form a functional ternary complex
Myeloperoxidase (MPO) and paraoxonase 1 (PON1) are high-density lipoprotein-associated (HDL-associated) proteins mechanistically linked to inflammation, oxidant stress, and atherosclerosis.MPO is a source of ROS during inflammation and can oxidize apolipoprotein A1 (APOA1) of HDL, impairing its atheroprotective functions.In contrast, PON1 fosters systemic antioxidant effects and promotes some of the atheroprotective properties attributed to HDL.Here, we demonstrate that MPO, PON1, and HDL bind to one another, forming a ternary complex, wherein PON1 partially inhibits MPO activity, while MPO inactivates PON1.MPO oxidizes PON1 on tyrosine 71 (Tyr 71 ), a modified residue found in human atheroma that is critical for HDL binding and PON1 function.Acute inflammation model studies with transgenic and knockout mice for either PON1 or MPO confirmed that MPO and PON1 reciprocally modulate each other's function in vivo.Further structure and function studies identified critical contact sites between APOA1 within HDL, PON1, and MPO, and proteomics studies of HDL recovered from acute coronary syndrome (ACS) subjects revealed enhanced chlorotyrosine content, site-specific PON1 methionine oxidation, and reduced PON1 activity.HDL thus serves as a scaffold upon which MPO and PON1 interact during inflammation, whereupon PON1 binding partially inhibits MPO activity, and MPO promotes site-specific oxidative modification and impairment of PON1 and APOA1 function.
DOI: 10.1002/ejhf.1697
2020
Cited 228 times
Evaluation of kidney function throughout the heart failure trajectory – a position statement from the Heart Failure Association of the European Society of Cardiology
Abstract Appropriate interpretation of changes in markers of kidney function is essential during the treatment of acute and chronic heart failure. Historically, kidney function was primarily assessed by serum creatinine and the calculation of estimated glomerular filtration rate. An increase in serum creatinine, also termed worsening renal function, commonly occurs in patients with heart failure, especially during acute heart failure episodes. Even though worsening renal function is associated with worse outcome on a population level, the interpretation of such changes within the appropriate clinical context helps to correctly assess risk and determine further treatment strategies. Additionally, it is becoming increasingly recognized that assessment of kidney function is more than just glomerular filtration rate alone. As such, a better evaluation of sodium and water handling by the renal tubules allows to determine the efficiency of loop diuretics (loop diuretic response and efficiency). Also, though neurohumoral blockers may induce modest deteriorations in glomerular filtration rate, their use is associated with improved long‐term outcome. Therefore, a better understanding of the role of cardio–renal interactions in heart failure in symptom development, disease progression and prognosis is essential. Indeed, perhaps even misinterpretation of kidney function is a leading cause of not attaining decongestion in acute heart failure and insufficient dosing of guideline‐directed medical therapy in general. This position paper of the Heart Failure Association Working Group on Cardio‐Renal Dysfunction aims at improving insights into the interpretation of renal function assessment in the different heart failure states, with the goal of improving heart failure care.
DOI: 10.1093/eurheartj/ehz056
2019
Cited 227 times
Deep learning for cardiovascular medicine: a practical primer
Abstract Deep learning (DL) is a branch of machine learning (ML) showing increasing promise in medicine, to assist in data classification, novel disease phenotyping and complex decision making. Deep learning is a form of ML typically implemented via multi-layered neural networks. Deep learning has accelerated by recent advances in computer hardware and algorithms and is increasingly applied in e-commerce, finance, and voice and image recognition to learn and classify complex datasets. The current medical literature shows both strengths and limitations of DL. Strengths of DL include its ability to automate medical image interpretation, enhance clinical decision-making, identify novel phenotypes, and select better treatment pathways in complex diseases. Deep learning may be well-suited to cardiovascular medicine in which haemodynamic and electrophysiological indices are increasingly captured on a continuous basis by wearable devices as well as image segmentation in cardiac imaging. However, DL also has significant weaknesses including difficulties in interpreting its models (the ‘black-box’ criticism), its need for extensive adjudicated (‘labelled’) data in training, lack of standardization in design, lack of data-efficiency in training, limited applicability to clinical trials, and other factors. Thus, the optimal clinical application of DL requires careful formulation of solvable problems, selection of most appropriate DL algorithms and data, and balanced interpretation of results. This review synthesizes the current state of DL for cardiovascular clinicians and investigators, and provides technical context to appreciate the promise, pitfalls, near-term challenges, and opportunities for this exciting new area.
DOI: 10.1161/circulationaha.108.844340
2010
Cited 226 times
Ventricular Geometry, Strain, and Rotational Mechanics in Pulmonary Hypertension
We tested the hypothesis that right ventricular (RV) pressure overload affects RV function and further influences left ventricular (LV) geometry, which adversely affects LV twist mechanics and segmental function.Echocardiographic images were prospectively acquired in 44 patients (age, 46+/-12 years; 82% women) with evidence of pulmonary hypertension (estimated pulmonary artery systolic pressure, 71+/-23 mm Hg) and in 44 age- and gender-matched healthy subjects. Patients with intrinsic LV diseases were excluded. RV lateral wall longitudinal strain (LS) and interventricular septal (IVS) LS were reduced in the pulmonary hypertension group compared with control subjects (-15.9+/-7.6% versus -25.5+/-6.1%, P<0.001; and -17.3+/-4.4% versus -20.2+/-3.9%, P=0.002, respectively), whereas LV lateral wall LS was preserved. RV lateral wall LS and IVS LS, but not LV lateral wall LS, correlated with pulmonary artery systolic pressure (r=0.56, P<0.01; r=0.32, P<0.01) and LV eccentricity index (r=0.57, P<0.01; r=0.57, P<0.01). IVS and LV lateral wall circumferential strain (CS) were both reduced in the pulmonary hypertension group. Although IVS CS and LV lateral wall CS correlated with pulmonary artery systolic pressure and LV eccentricity index, after adjustment of CS for LV eccentricity index, differences between groups persisted for IVS CS (P<0.01) but not LV lateral wall CS (P=0.09). LV torsion was decreased in patients with pulmonary hypertension compared with control subjects (9.6+/-4.9 degrees versus 14.7+/-4.9 degrees , P<0.001). LV torsion inversely correlated with pulmonary artery systolic pressure (r=-0.39, P<0.01) and LV eccentricity index (r=-0.3, P<0.01). LV untwisting rates were similar in both groups (P=0.7).Chronic RV pressure overload directly affects RV longitudinal systolic deformation. RV pressure overload further influences IVS and LV geometry, which impairs LV torsion and segmental LS and CS, more for the IVS than for the free wall of the LV.
DOI: 10.1016/j.jacc.2009.07.037
2009
Cited 224 times
Role of the CHADS2Score in the Evaluation of Thromboembolic Risk in Patients With Atrial Fibrillation Undergoing Transesophageal Echocardiography Before Pulmonary Vein Isolation
The goals of this study were to determine: 1) if low-risk patients assessed by a CHADS2score, a clinical scoring system quantifying a risk of stroke in patients with atrial fibrillation (AF), require a routine screening transesophageal echocardiogram (TEE) before pulmonary vein isolation (PVI); and 2) the relationship of a CHADS2score with left atrial (LA)/left atrial appendage (LAA) spontaneous echo contrast, sludge, and thrombus. There is no clear consensus of whether a screening TEE before catheter ablation of AF should be performed in every patient. Initial TEEs for pre-PVI of 1,058 AF patients (age 57 ± 11 years, 80% men) were reviewed and compared with a CHADS2score. CHADS2scores of 0, 1, 2, 3, 4, 5, and 6 were present in 47%, 33%, 14%, 5%, 1%, 0.3%, and 0% of patients, respectively. The prevalence of LA/LAA thrombus, sludge, and spontaneous echo contrast were present in 0.6%, 1.5%, and 35%. The prevalence of LA/LAA thrombus/sludge increased with ascending CHADS2score (scores 0 [0%], 1 [2%], 2 [5%], 3 [9%], and 4 to 6 [11%], p < 0.01). No patient with a CHADS2score of 0 had LA/LAA sludge/thrombus. In a multivariate model, history of congestive heart failure and left ventricular ejection fraction <35% were significantly associated with sludge/thrombus. The prevalence of LA/LAA sludge/thrombus in patients with AF undergoing a pre-PVI screening TEE is very low (<2%) and increases significantly with higher CHADS2scores. This suggests that a screening TEE before PVI should be performed in patients with a CHADS2score of ≥1, and in patients with a CHADS2score of 0 when the AF is persistent and therapeutic anticoagulation has not been maintained for 4 weeks before the procedure.
DOI: 10.1016/j.ygeno.2014.12.002
2015
Cited 221 times
RNA-Seq identifies novel myocardial gene expression signatures of heart failure
Heart failure is a complex clinical syndrome and has become the most common reason for adult hospitalization in developed countries. Two subtypes of heart failure, ischemic heart disease (ISCH) and dilated cardiomyopathy (DCM), have been studied using microarray platforms. However, microarray has limited resolution. Here we applied RNA sequencing (RNA-Seq) to identify gene signatures for heart failure from six individuals, including three controls, one ISCH and two DCM patients. Using genes identified from this small RNA-Seq dataset, we were able to accurately classify heart failure status in a much larger set of 313 individuals. The identified genes significantly overlapped with genes identified via genome-wide association studies for cardiometabolic traits and the promoters of those genes were enriched for binding sites for transcriptions factors. Our results indicate that it is possible to use RNA-Seq to classify disease status for complex diseases such as heart failure using an extremely small training dataset.
DOI: 10.1016/j.jacc.2018.07.092
2018
Cited 218 times
Tenosynovial and Cardiac Amyloidosis in Patients Undergoing Carpal Tunnel Release
Patients with cardiac amyloidosis often have carpal tunnel syndrome that precedes cardiac manifestations by several years. However, the prevalence of cardiac involvement at the time of carpal tunnel surgery has not been established. The authors sought to identify the prevalence and type of amyloid deposits in patients undergoing carpal tunnel surgery and evaluate for cardiac involvement. The authors also sought to determine if patients with soft tissue transthyretin (TTR) amyloid had abnormal TTR tetramer kinetic stability. This was a prospective, cross-sectional, multidisciplinary study of consecutive men age ≥50 years and women ≥60 years undergoing carpal tunnel release surgery. Biopsy specimens of tenosynovial tissue were obtained and stained with Congo red; those with confirmed amyloid deposits were typed with mass spectrometry and further evaluated for cardiac involvement with biomarkers, electrocardiography, echocardiography with longitudinal strain, and technetium pyrophosphate scintigraphy. Additionally, serum TTR concentration and tetramer kinetic stability were examined. Of 98 patients enrolled (median age 68 years, 51% male), 10 (10.2%) had a positive biopsy for amyloid (7 ATTR, 2 light chain [AL], 1 untyped). Two patients were diagnosed with hereditary ATTR (Leu58His and Ala81Thr), 2 were found to have cardiac involvement (1 AL, 1 ATTR wild-type), and 3 were initiated on therapy. In those patients who had biopsy-diagnosed ATTR, there was no difference in plasma TTR concentration or tetramer kinetic stability. In a cohort of patients undergoing carpal tunnel release surgery, Congo red staining of tenosynovial tissue detected amyloid deposits in 10.2% of patients. Concomitant cardiac evaluation identified patients with involvement of the myocardium, allowing for implementation of disease-modifying therapy. (Carpal Tunnel Syndrome and Amyloid Cardiomyopathy; NCT02792790)
DOI: 10.1172/jci94601
2018
Cited 216 times
l-Carnitine in omnivorous diets induces an atherogenic gut microbial pathway in humans
BACKGROUND. l-Carnitine, an abundant nutrient in red meat, accelerates atherosclerosis in mice via gut microbiotadependent formation of trimethylamine (TMA) and trimethylamine N-oxide (TMAO) via a multistep pathway involving an atherogenic intermediate, γ-butyrobetaine (γBB).The contribution of γBB in gut microbiota-dependent l-carnitine metabolism in humans is unknown. METHODS.Omnivores and vegans/vegetarians ingested deuterium-labeled l-carnitine (d 3 -l-carnitine) or γBB (d 9 -γBB), and both plasma metabolites and fecal polymicrobial transformations were examined at baseline, following oral antibiotics, or following chronic (≥2 months) l-carnitine supplementation.Human fecal commensals capable of performing each step of the l-carnitine→γBB→TMA transformation were identified. RESULTS.Studies with oral d 3 -l-carnitine or d 9 -γBB before versus after antibiotic exposure revealed gut microbiota contribution to the initial 2 steps in a metaorganismal l-carnitine →γBB→TMA→TMAO pathway in subjects.Moreover, a striking increase in d 3 -TMAO generation was observed in omnivores over vegans/vegetarians (>20-fold; P = 0.001) following oral d 3 -l-carnitine ingestion, whereas fasting endogenous plasma l-carnitine and γBB levels were similar in vegans/ vegetarians (n = 32) versus omnivores (n = 40).Fecal metabolic transformation studies, and oral isotope tracer studies before versus after chronic l-carnitine supplementation, revealed that omnivores and vegans/vegetarians alike rapidly converted carnitine to γBB, whereas the second gut microbial transformation, γBB→TMA, was diet inducible (l-carnitine, omnivorous).Extensive anaerobic subculturing of human feces identified no single commensal capable of l-carnitine→TMA transformation, multiple community members that converted l-carnitine to γBB, and only 1 Clostridiales bacterium, Emergencia timonensis, that converted γBB to TMA.In coculture, E. timonensis promoted the complete l-carnitine→TMA transformation. CONCLUSION.In humans, dietary l-carnitine is converted into the atherosclerosis-and thrombosis-promoting metabolite TMAO via 2 sequential gut microbiota-dependent transformations: (a) initial rapid generation of the atherogenic intermediate γBB, followed by (b) transformation into TMA via low-abundance microbiota in omnivores, and to a markedly lower extent, in vegans/vegetarians.Gut microbiota γBB→TMA/TMAO transformation is induced by omnivorous dietary patterns and chronic l-carnitine exposure.
DOI: 10.1161/circheartfailure.113.000895
2014
Cited 213 times
Loop Diuretic Efficiency
Background— Rather than the absolute dose of diuretic or urine output, the primary signal of interest when evaluating diuretic responsiveness is the efficiency with which the kidneys can produce urine after a given dose of diuretic. As a result, we hypothesized that a metric of diuretic efficiency (DE) would capture distinct prognostic information beyond that of raw fluid output or diuretic dose. Methods and Results— We independently analyzed 2 cohorts: (1) consecutive admissions at the University of Pennsylvania (Penn) with a primary discharge diagnosis of heart failure (n=657) and (2) patients in the Evaluation Study of Congestive Heart Failure and Pulmonary Artery Catheterization Effectiveness (ESCAPE) data set (n=390). DE was estimated as the net fluid output produced per 40 mg of furosemide equivalents, then dichotomized into high versus low DE based on the median value. There was only a moderate correlation between DE and both intravenous diuretic dose and net fluid output (r 2 ≤0.26 for all comparisons), indicating that DE was describing unique information. With the exception of metrics of renal function and preadmission diuretic therapy, traditional baseline characteristics, including right heart catheterization variables, were not consistently associated with DE. Low DE was associated with worsened survival even after adjusting for in-hospital diuretic dose, fluid output, in addition to baseline characteristics (Penn: hazards ratio [HR], 1.36; 95% confidence interval [CI], 1.04−1.78; P =0.02; ESCAPE: HR, 2.86; 95% CI, 1.53−5.36; P =0.001). Conclusions— Although in need of validation in less-selected populations, low DE during decongestive therapy portends poorer long-term outcomes above and beyond traditional prognostic factors in patients hospitalized with decompensated heart failure.
DOI: 10.1161/circulationaha.116.025338
2017
Cited 206 times
Gut Microbe-Generated Trimethylamine <i>N</i> -Oxide From Dietary Choline Is Prothrombotic in Subjects
HomeCirculationVol. 135, No. 17Gut Microbe-Generated Trimethylamine N-Oxide From Dietary Choline Is Prothrombotic in Subjects Free AccessLetterPDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toFree AccessLetterPDF/EPUBGut Microbe-Generated Trimethylamine N-Oxide From Dietary Choline Is Prothrombotic in Subjects Weifei Zhu, PhD, Zeneng Wang, PhD, W. H. Wilson Tang, MD and Stanley L. Hazen, MD, PhD Weifei ZhuWeifei Zhu From Department of Cellular and Molecular Medicine, Lerner Research Institute (W.Z., Z.W., W.H.W.T., S.L.H.), Center for Microbiome & Human Health (W.Z., Z.W., W.H.W.T., S.L.H.), and Heart and Vascular Institute (W.H.W.T., S.L.H.), Cleveland Clinic, OH. Search for more papers by this author , Zeneng WangZeneng Wang From Department of Cellular and Molecular Medicine, Lerner Research Institute (W.Z., Z.W., W.H.W.T., S.L.H.), Center for Microbiome & Human Health (W.Z., Z.W., W.H.W.T., S.L.H.), and Heart and Vascular Institute (W.H.W.T., S.L.H.), Cleveland Clinic, OH. Search for more papers by this author , W. H. Wilson TangW. H. Wilson Tang From Department of Cellular and Molecular Medicine, Lerner Research Institute (W.Z., Z.W., W.H.W.T., S.L.H.), Center for Microbiome & Human Health (W.Z., Z.W., W.H.W.T., S.L.H.), and Heart and Vascular Institute (W.H.W.T., S.L.H.), Cleveland Clinic, OH. Search for more papers by this author and Stanley L. HazenStanley L. Hazen From Department of Cellular and Molecular Medicine, Lerner Research Institute (W.Z., Z.W., W.H.W.T., S.L.H.), Center for Microbiome & Human Health (W.Z., Z.W., W.H.W.T., S.L.H.), and Heart and Vascular Institute (W.H.W.T., S.L.H.), Cleveland Clinic, OH. Search for more papers by this author Originally published25 Apr 2017https://doi.org/10.1161/CIRCULATIONAHA.116.025338Circulation. 2017;135:1671–1673We previously showed gut microbial production of trimethylamine N-oxide (TMAO) from dietary nutrients like choline, lecithin, and L-carnitine is linked to the development of cardiovascular diseases.1–3 We also recently reported that plasma TMAO levels are associated with incident thrombotic event risk in subjects, and that TMAO both enhances platelet responsiveness to multiple agonists by augmenting stimulus-dependent Ca2+ signaling and heightens thrombosis potential in animal models.4 Specifically, a role for TMAO and gut microbiota in transmitting heightened thrombosis potential in vivo was supported by both direct TMAO infusion and microbial transplantation studies.4 A Western diet, rich in choline, is associated with heightened thrombosis risk; however, the effect of dietary choline on TMAO and platelet hyperresponsiveness in human subjects has not yet been reported.We prospectively recruited healthy vegans/vegetarians (n=8) and omnivores (n=10) with no preceding (1-month) history of antibiotics or probiotics. This single-center study was approved by the Cleveland Clinic Institutional Review Board. After informed consent, subjects (46±5 years of age, 40% male, nonsmokers without hypertension, diabetes mellitus, or cardiovascular disease) were given oral choline supplementation (choline bitartrate 500 mg twice daily, ≈450 mg total choline/day) for 2 months with monthly blood testing after overnight fast. Both vegan/vegetarian and omnivore alike showed significant >10-fold increases in plasma TMAO levels at both 1- and 2-month periods (P<0.01 each; Figure, A), with corresponding enhanced platelet aggregation responses to submaximal adenosine diphosphate (5µM) after choline supplementation (Figure, A). Moreover, a striking dose-dependent association was observed between plasma TMAO levels and platelet function (Figure, B). Similarly, among all subjects in the study, a significant association was noted between change from baseline in TMAO level and change from baseline in platelet aggregation (Spearman rho=0.38, P=0.03).Download figureDownload PowerPointFigure. Oral choline supplementation increases fasting trimethylamine N-oxide (TMAO) levels, enhances platelet aggregation, and attenuates the antiplatelet effect observed with aspirin.A, Plasma TMAO levels and platelet aggregation in response to submaximal adenosine diphosphate (5µM) in vegan/vegetarian and omnivore groups. B, Correlation between plasma TMAO and platelet aggregation responses among the indicated groups. Spearman correlations and P values shown. C, Effect of choline supplementation on TMAO and platelet aggregation responses in omnivores in the absence versus presence of aspirin (ASA). All data shown are mean (±SEM) with the indicated number of subjects. Asterisks shown represent P<0.05 for comparison of aggregation responses off versus on ASA for the corresponding time point. P values were calculated with Wilcoxon rank sum test for two-group comparisons and Wilcoxon signed rank test for pairwise comparisons.We next tested whether platelet hyperresponsiveness associated with choline supplementation and elevated TMAO was observed in the presence of aspirin. Omnivores previously examined in the absence of aspirin had a choline supplement-free washout period of at least 1 month and then were started on aspirin (81 mg each evening) for 1 month before a baseline evaluation, followed by 2 months of choline supplementation. Compared with baseline, choline again increased both fasting plasma TMAO levels and adenosine diphosphate-dependent platelet aggregation responses at 1 and 2 months of supplementation; however, both the degree of TMAO elevation and platelet hyperresponsiveness were attenuated by aspirin therapy (Figure, C).These studies show for the first time a direct prothrombotic effect of dietary choline and elevated levels of the gut microbial metabolite TMAO in humans. They also suggest the platelet hyperresponsiveness mediated by elevated TMAO can be attenuated by a low dose of aspirin. It is important to note that they suggest elevated levels of the gut microbe-generated metabolite TMAO may overcome the antiplatelet effects of low-dose aspirin—a hypothesis that warrants further investigation, particularly in subjects at high cardiovascular risk. An unanticipated finding was that low-dose aspirin partially reduced choline supplement-dependent rise in TMAO. Although the mechanism for this result is unknown, aspirin has been reported to alter the composition of the gut microbial community.5 Finally, aspirin use in primary prevention subjects has recently been debated. The present studies, coupled with published studies linking heightened TMAO levels with thrombotic event risk,4 suggest studies are warranted to explore if low-dose aspirin is beneficial among subjects with elevated TMAO and no clear contraindications to aspirin.Weifei Zhu, PhDZeneng Wang, PhDW. H. Wilson Tang, MDStanley L. Hazen, MD, PhDSources of FundingThis research was supported by grants from the National Institutes of Health and the Office of Dietary Supplements (R01HL103866, R01DK106000, R01HL126827).DisclosuresDrs Hazen and Wang are named as coinventors on pending and issued patents held by the Cleveland Clinic relating to cardiovascular diagnostics and therapeutics. Dr Hazen is a paid consultant for Esperion and P&G; has received research funds from P&G, Pfizer Inc., Roche Diagnostics, and Takeda; and also reports he may receive royalty payments for inventions or discoveries related to cardiovascular diagnostics or therapeutics from P&G, Cleveland HeartLab, Siemens, Esperion, and Frantz Biomarkers, LLC. Dr Wang reports he may receive royalty payments for inventions or discoveries related to cardiovascular diagnostics or therapeutics from Cleveland HeartLab. The other authors report no conflicts of interest.FootnotesCirculation is available at http://circ.ahajournals.org.Correspondence to: Stanley L. Hazen, MD, PhD, Department of Cellular & Molecular Medicine, Cleveland Clinic, 9500 Euclid Ave, NC-10, Cleveland, OH 44195. E-mail [email protected]cf.orgReferences1. Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt EB, Fu X, Chung YM, Wu Y, Schauer P, Smith JD, Allayee H, Tang WH, DiDonato JA, Lusis AJ, Hazen SL. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease.Nature. 2011; 472:57–63. doi: 10.1038/nature09922.CrossrefMedlineGoogle Scholar2. Tang WH, Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, Wu Y, Hazen SL. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk.N Engl J Med. 2013; 368:1575–1584. doi: 10.1056/NEJMoa1109400.CrossrefMedlineGoogle Scholar3. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X, Wu Y, Li L, Smith JD, DiDonato JA, Chen J, Li H, Wu GD, Lewis JD, Warrier M, Brown JM, Krauss RM, Tang WH, Bushman FD, Lusis AJ, Hazen SL. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis.Nat Med. 2013; 19:576–585. doi: 10.1038/nm.3145.CrossrefMedlineGoogle Scholar4. Zhu W, Gregory JC, Org E, Buffa JA, Gupta N, Wang Z, Li L, Fu X, Wu Y, Mehrabian M, Sartor RB, McIntyre TM, Silverstein RL, Tang WH, DiDonato JA, Brown JM, Lusis AJ, Hazen SL. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk.Cell. 2016; 165:111–124. doi: 10.1016/j.cell.2016.02.011.CrossrefMedlineGoogle Scholar5. Rogers MA, Aronoff DM. The influence of non-steroidal anti-inflammatory drugs on the gut microbiome.Clin Microbiol Infect. 2016; 22:178.e1–178.e9. doi: 10.1016/j.cmi.2015.10.003.CrossrefGoogle Scholar Previous Back to top Next FiguresReferencesRelatedDetailsCited By Panyod S, Wu W, Chen P, Chong K, Yang Y, Chuang H, Chen C, Chen R, Liu P, Chung C, Huang H, Lin A, Shen T, Yang K, Huang T, Hsu C, Ho C, Kao H, Orekhov A, Wu M and Sheen L (2022) Atherosclerosis amelioration by allicin in raw garlic through gut microbiota and trimethylamine-N-oxide modulation, npj Biofilms and Microbiomes, 10.1038/s41522-022-00266-3, 8:1, Online publication date: 1-Dec-2022. Xue J, Xu J, Zhao M, Jin A, Cheng A, Jiang X, Li K, Lin J, Meng X, Li H, Zheng L and Wang Y (2022) Residual Risk of Trimethylamine‐N‐Oxide and Choline for Stroke Recurrence in Patients With Intensive Secondary Therapy, Journal of the American Heart Association, 11:19, Online publication date: 4-Oct-2022.Wang M, Wang Z, Lee Y, Lai H, de Oliveira Otto M, Lemaitre R, Fretts A, Sotoodehnia N, Budoff M, DiDonato J, McKnight B, Tang W, Psaty B, Siscovick D, Hazen S and Mozaffarian D (2022) Dietary Meat, Trimethylamine N-Oxide-Related Metabolites, and Incident Cardiovascular Disease Among Older Adults: The Cardiovascular Health Study, Arteriosclerosis, Thrombosis, and Vascular Biology, 42:9, (e273-e288), Online publication date: 1-Sep-2022. Voigt R, Wang Z, Brown J, Engen P, Naqib A, Goetz C, Hall D, Metman L, Shaikh M, Forsyth C and Keshavarzian A (2022) Gut microbial metabolites in Parkinson's disease: Association with lifestyle, disease characteristics, and treatment status, Neurobiology of Disease, 10.1016/j.nbd.2022.105780, 170, (105780), Online publication date: 1-Aug-2022. Rahman M, Islam F, -Or-Rashid M, Mamun A, Rahaman M, Islam M, Meem A, Sutradhar P, Mitra S, Mimi A, Emran T, Fatimawali , Idroes R, Tallei T, Ahmed M and Cavalu S (2022) The Gut Microbiota (Microbiome) in Cardiovascular Disease and Its Therapeutic Regulation, Frontiers in Cellular and Infection Microbiology, 10.3389/fcimb.2022.903570, 12 Liu W, Zhang X, Wu Z, Huang K, Yang C and Yang L (2022) Brain–heart communication in health and diseases, Brain Research Bulletin, 10.1016/j.brainresbull.2022.02.012, 183, (27-37), Online publication date: 1-Jun-2022. Amrein M, Li X, Walter J, Wang Z, Zimmermann T, Strebel I, Honegger U, Leu K, Schäfer I, Twerenbold R, Puelacher C, Glarner N, Nestelberger T, Koechlin L, Ceresa B, Haaf P, Bakula A, Zellweger M, Hazen S and Mueller C (2022) Gut microbiota-dependent metabolite trimethylamine N-oxide (TMAO) and cardiovascular risk in patients with suspected functionally relevant coronary artery disease (fCAD), Clinical Research in Cardiology, 10.1007/s00392-022-01992-6, 111:6, (692-704), Online publication date: 1-Jun-2022. Mu F, Tang M, Guan Y, Lin R, Zhao M, Zhao J, Huang S, Zhang H, Wang J and Tang H (2022) Knowledge Mapping of the Links Between the Gut Microbiota and Heart Failure: A Scientometric Investigation (2006–2021), Frontiers in Cardiovascular Medicine, 10.3389/fcvm.2022.882660, 9 Tousoulis D, Guzik T, Padro T, Duncker D, De Luca G, Eringa E, Vavlukis M, Antonopoulos A, Katsimichas T, Cenko E, Djordjevic-Dikic A, Fleming I, Manfrini O, Trifunovic D, Antoniades C and Crea F (2022) Mechanisms, therapeutic implications, and methodological challenges of gut microbiota and cardiovascular diseases: a position paper by the ESC Working Group on Coronary Pathophysiology and Microcirculation, Cardiovascular Research, 10.1093/cvr/cvac057 Thomas M, DiBella M, Blesso C, Malysheva O, Caudill M, Sholola M, Cooperstone J and Fernandez M (2022) Comparison between Egg Intake versus Choline Supplementation on Gut Microbiota and Plasma Carotenoids in Subjects with Metabolic Syndrome, Nutrients, 10.3390/nu14061179, 14:6, (1179) Canyelles M, Plaza M, Rotllan N, Llobet D, Julve J, Mojal S, Diaz-Ricart M, Soria J, Escolà-Gil J, Tondo M, Blanco-Vaca F and Souto J (2022) TMAO and Gut Microbial-Derived Metabolites TML and γBB Are Not Associated with Thrombotic Risk in Patients with Venous Thromboembolism, Journal of Clinical Medicine, 10.3390/jcm11051425, 11:5, (1425) Wang Z, Tang W, O’Connell T, Garcia E, Jeyarajah E, Li X, Jia X, Weeks T and Hazen S (2022) Circulating trimethylamine N-oxide levels following fish or seafood consumption, European Journal of Nutrition, 10.1007/s00394-022-02803-4 Helsley R, Miyata T, Kadam A, Varadharajan V, Sangwan N, Huang E, Banerjee R, Brown A, Fung K, Massey W, Neumann C, Orabi D, Osborn L, Schugar R, McMullen M, Bellar A, Poulsen K, Kim A, Pathak V, Mrdjen M, Anderson J, Willard B, McClain C, Mitchell M, McCullough A, Radaeva S, Barton B, Szabo G, Dasarathy S, Garcia-Garcia J, Rotroff D, Allende D, Wang Z, Hazen S, Nagy L and Brown J (2022) Gut microbial trimethylamine is elevated in alcohol-associated hepatitis and contributes to ethanol-induced liver injury in mice, eLife, 10.7554/eLife.76554, 11 Deidda M, Noto A, Cadeddu Dessalvi C, Andreini D, Andreotti F, Ferrannini E, Latini R, Maggioni A, Magnoni M and Mercuro G (2022) Why Do High-Risk Patients Develop or Not Develop Coronary Artery Disease? Metabolic Insights from the CAPIRE Study, Metabolites, 10.3390/metabo12020123, 12:2, (123) Schugar R, Gliniak C, Osborn L, Massey W, Sangwan N, Horak A, Banerjee R, Orabi D, Helsley R, Brown A, Burrows A, Finney C, Fung K, Allen F, Ferguson D, Gromovsky A, Neumann C, Cook K, McMillan A, Buffa J, Anderson J, Mehrabian M, Goudarzi M, Willard B, Mak T, Armstrong A, Swanson G, Keshavarzian A, Garcia-Garcia J, Wang Z, Lusis A, Hazen S and Brown J (2022) Gut microbe-targeted choline trimethylamine lyase inhibition improves obesity via rewiring of host circadian rhythms, eLife, 10.7554/eLife.63998, 11 Li Z, Hui J, Li S, Cao T, Zhang J, Mao X, Wang F, Wang F, He P, You Y and Xi G (2022) Trimethylamine N ‐oxide predicts stroke severity in diabetic patients with acute ischaemic stroke and is related to glycemic variability , European Journal of Neurology, 10.1111/ene.15249 Zou Y, Song X, Liu N, Sun W and Liu B (2022) Intestinal Flora: A Potential New Regulator of Cardiovascular Disease, Aging and disease, 10.14336/AD.2021.1022, 13:3, (753), . Aganetti M, Cruz C, Galvão I, Engels D, Ricci M and Vieira A (2022) The Gut Microbiota and Immunopathophysiology Comprehensive Pharmacology, 10.1016/B978-0-12-820472-6.00128-6, (492-514), . Gasmi A, Noor S, Piscopo S and Berthouze S (2022) Gut Microbial Dysbiosis and Cardiovascular Diseases Comprehensive Gut Microbiota, 10.1016/B978-0-12-819265-8.00050-4, (455-474), . Gabbianelli R and Bordoni L (2022) Trimethylamine N-Oxide (TMAO) as a Biomarker Biomarkers in Nutrition, 10.1007/978-3-030-81304-8_2-1, (1-20), . Krueger E, Beales J, Russon K, Elison W, Davis J, Hansen J, Neilson A, Hansen J and Tessem J (2021) Gut Metabolite Trimethylamine N-Oxide Protects INS-1 β-Cell and Rat Islet Function under Diabetic Glucolipotoxic Conditions, Biomolecules, 10.3390/biom11121892, 11:12, (1892) Wang B, Qiu J, Lian J, Yang X and Zhou J (2021) Gut Metabolite Trimethylamine-N-Oxide in Atherosclerosis: From Mechanism to Therapy, Frontiers in Cardiovascular Medicine, 10.3389/fcvm.2021.723886, 8 Ferrell M, Bazeley P, Wang Z, Levison B, Li X, Jia X, Krauss R, Knight R, Lusis A, Garcia‐Garcia J, Hazen S and Tang W (2021) Fecal Microbiome Composition Does Not Predict Diet‐Induced TMAO Production in Healthy Adults, Journal of the American Heart Association, 10:21, Online publication date: 2-Nov-2021. Bishai J and Palm N (2021) Small Molecule Metabolites at the Host–Microbiota Interface, The Journal of Immunology, 10.4049/jimmunol.2100528, 207:7, (1725-1733), Online publication date: 1-Oct-2021. Williams K, Fughhi I, Fugar S, Mazur M, Gates S, Sawyer S, Patel H, Chambers D, McDaniel R, Reiser J and Mason T (2021) Nutrition Intervention for Reduction of Cardiovascular Risk in African Americans Using the 2019 American College of Cardiology/American Heart Association Primary Prevention Guidelines, Nutrients, 10.3390/nu13103422, 13:10, (3422) McGranaghan P, Kirwan J, Garcia-Rivera M, Pieske B, Edelmann F, Blaschke F, Appunni S, Saxena A, Rubens M, Veledar E and Trippel T (2021) Lipid Metabolite Biomarkers in Cardiovascular Disease: Discovery and Biomechanism Translation from Human Studies, Metabolites, 10.3390/metabo11090621, 11:9, (621) NERGİZ-UNAL R and GÖNEN B (2021) Kardiyovasküler Hastalıklarda Bağırsak Mikrobiyota Metaboliti Trimetilamin N-oksit (TMAO) : Önleme ve Tedavi İçin Yeni Bir Molekül mü?, Akdeniz Medical Journal, 10.53394/akd.982129, (436-447) Panyod S, Wu W, Chen C, Wu M, Ho C and Sheen L (2021) Modulation of gut microbiota by foods and herbs to prevent cardiovascular diseases, Journal of Traditional and Complementary Medicine, 10.1016/j.jtcme.2021.09.006, Online publication date: 1-Sep-2021. Wilcox J, Skye S, Graham B, Zabell A, Li X, Li L, Shelkay S, Fu X, Neale S, O'Laughlin C, Peterson K, Hazen S and Tang W (2021) Dietary Choline Supplements, but Not Eggs, Raise Fasting TMAO Levels in Participants with Normal Renal Function: A Randomized Clinical Trial, The American Journal of Medicine, 10.1016/j.amjmed.2021.03.016, 134:9, (1160-1169.e3), Online publication date: 1-Sep-2021. Krueger E, Lloyd T and Tessem J (2021) The Accumulation and Molecular Effects of Trimethylamine N-Oxide on Metabolic Tissues: It’s Not All Bad, Nutrients, 10.3390/nu13082873, 13:8, (2873) Witkowski M, Witkowski M, Friebel J, Buffa J, Li X, Wang Z, Sangwan N, Li L, DiDonato J, Tizian C, Haghikia A, Kirchhofer D, Mach F, Räber L, Matter C, Tang W, Landmesser U, Lüscher T, Rauch U and Hazen S (2021) Vascular endothelial tissue factor contributes to trimethylamine N-oxide-enhanced arterial thrombosis, Cardiovascular Research, 10.1093/cvr/cvab263 Lemaitre R, Jensen P, Wang Z, Fretts A, McKnight B, Nemet I, Biggs M, Sotoodehnia N, de Oliveira Otto M, Psaty B, Siscovick D, Hazen S and Mozaffarian D (2021) Association of Trimethylamine N -Oxide and Related Metabolites in Plasma and Incident Type 2 Diabetes , JAMA Network Open, 10.1001/jamanetworkopen.2021.22844, 4:8, (e2122844) Franczyk B, Gluba-Brzózka A, Ławiński J, Rysz-Górzyńska M and Rysz J (2021) Metabolomic Profile in Venous Thromboembolism (VTE), Metabolites, 10.3390/metabo11080495, 11:8, (495) Szabo Z, Koczka V, Marosvolgyi T, Szabo E, Frank E, Polyak E, Fekete K, Erdelyi A, Verzar Z and Figler M (2021) Possible Biochemical Processes Underlying the Positive Health Effects of Plant-Based Diets—A Narrative Review, Nutrients, 10.3390/nu13082593, 13:8, (2593) Anselmi G, Gagliardi L, Egidi G, Leone S, Gasbarrini A, Miggiano G and Galiuto L (2020) Gut Microbiota and Cardiovascular Diseases, Cardiology in Review, 10.1097/CRD.0000000000000327, 29:4, (195-204), Online publication date: 1-Jul-2021. Zhu W, Romano K, Li L, Buffa J, Sangwan N, Prakash P, Tittle A, Li X, Fu X, Androjna C, DiDonato A, Brinson K, Trapp B, Fischbach M, Rey F, Hajjar A, DiDonato J and Hazen S (2021) Gut microbes impact stroke severity via the trimethylamine N-oxide pathway, Cell Host & Microbe, 10.1016/j.chom.2021.05.002, 29:7, (1199-1208.e5), Online publication date: 1-Jul-2021. Ding B, Peterzan M, Mózes F, Rider O, Valkovič L and Rodgers C (2021) Water‐suppression cycling 3‐T cardiac 1 H‐MRS detects altered creatine and choline in patients with aortic or mitral stenosis , NMR in Biomedicine, 10.1002/nbm.4513, 34:7, Online publication date: 1-Jul-2021. Naghipour S, Cox A, Peart J, Du Toit E and Headrick J (2020) Trimethylamine N -oxide: heart of the microbiota–CVD nexus? , Nutrition Research Reviews, 10.1017/S0954422420000177, 34:1, (125-146), Online publication date: 1-Jun-2021. Tang W, Li X, Wu Y, Wang Z, Khaw K, Wareham N, Nieuwdorp M, Boekholdt S and Hazen S (2021) Plasma trimethylamine N-oxide (TMAO) levels predict future risk of coronary artery disease in apparently healthy individuals in the EPIC-Norfolk prospective population study, American Heart Journal, 10.1016/j.ahj.2021.01.020, 236, (80-86), Online publication date: 1-Jun-2021. Iglesias-Carres L, Hughes M, Steele C, Ponder M, Davy K and Neilson A (2021) Use of dietary phytochemicals for inhibition of trimethylamine N-oxide formation, The Journal of Nutritional Biochemistry, 10.1016/j.jnutbio.2021.108600, 91, (108600), Online publication date: 1-May-2021. Jansen V, Gerdes V, Middeldorp S and van Mens T (2021) Gut microbiota and their metabolites in cardiovascular disease, Best Practice & Research Clinical Endocrinology & Metabolism, 10.1016/j.beem.2021.101492, 35:3, (101492), Online publication date: 1-May-2021. Li X, Su C, Jiang Z, Yang Y, Zhang Y, Yang M, Zhang X, Du Y, Zhang J, Wang L, Jiang J and Hong B (2021) Berberine attenuates choline-induced atherosclerosis by inhibiting trimethylamine and trimethylamine-N-oxide production via manipulating the gut microbiome, npj Biofilms and Microbiomes, 10.1038/s41522-021-00205-8, 7:1 Fryc J and Naumnik B (2021) Thrombolome and Its Emerging Role in Chronic Kidney Diseases, Toxins, 10.3390/toxins13030223, 13:3, (223) Mamic P, Chaikijurajai T and Tang W (2021) Gut microbiome - A potential mediator of pathogenesis in heart failure and its comorbidities: State-of-the-art review, Journal of Molecular and Cellular Cardiology, 10.1016/j.yjmcc.2020.12.001, 152, (105-117), Online publication date: 1-Mar-2021. Wu Y, Zhu B, Chen Z, Duan J, Luo A, Yang L and Yang C (2021) New Insights Into the Comorbidity of Coronary Heart Disease and Depression, Current Problems in Cardiology, 10.1016/j.cpcardiol.2019.03.002, 46:3, (100413), Online publication date: 1-Mar-2021. Massey W and Brown J (2020) The Gut Microbial Endocrine Organ in Type 2 Diabetes, Endocrinology, 10.1210/endocr/bqaa235, 162:2, Online publication date: 1-Feb-2021. Rajendiran E, Ramadass B and Ramprasath V (2021) Understanding connections and roles of gut microbiome in cardiovascular diseases, Canadian Journal of Microbiology, 10.1139/cjm-2020-0043, 67:2, (101-111), Online publication date: 1-Feb-2021. Zhang Y, Wang Y, Ke B and Du J (2021) TMAO: how gut microbiota contributes to heart failure, Translational Research, 10.1016/j.trsl.2020.08.007, 228, (109-125), Online publication date: 1-Feb-2021. Castiglione Morelli M, Ostuni A, Giangaspero B, Cecchini S, Carluccio A and Boni R (2021) Relationships between Seminal Plasma Metabolites, Semen Characteristics and Sperm Kinetics in Donkey (Equus asinus), Animals, 10.3390/ani11010201, 11:1, (201) Andraos S, Jones B, Lange K, Clifford S, Thorstensen E, Kerr J, Wake M, Saffery R, Burgner D and O'Sullivan J (2020) Trimethylamine N-oxide (TMAO) Is not Associated with Cardiometabolic Phenotypes and Inflammatory Markers in Children and Adults, Current Developments in Nutrition, 10.1093/cdn/nzaa179, 5:1, Online publication date: 15-Jan-2021. Xu J and Yang Y (2021) Gut microbiome and its meta-omics perspectives: profound implications for cardiovascular diseases, Gut Microbes, 10.1080/19490976.2021.1936379, 13:1, Online publication date: 1-Jan-2021. Zhao X, Oduro P, Tong W, Wang Y, Gao X and Wang Q (2021) Therapeutic potential of natural products against atherosclerosis: Targeting on gut microbiota, Pharmacological Research, 10.1016/j.phrs.2020.105362, 163, (105362), Online publication date: 1-Jan-2021. Guasti L, Galliazzo S, Molaro M, Visconti E, Pennella B, Gaudio G, Lupi A, Grandi A and Squizzato A (2020) TMAO as a biomarker of cardiovascular events: a systematic review and meta-analysis, Internal and Emergency Medicine, 10.1007/s11739-020-02470-5, 16:1, (201-207), Online publication date: 1-Jan-2021. Chaikijurajai T, Wilcox J and Tang W (2021) Role of the Microbiome in Cardiovascular Disease Prevention and Treatment of Cardiovascular Disease, 10.1007/978-3-030-78177-4_15, (225-250), . Zhu S, Jiang Y, Xu K, Cui M, Ye W, Zhao G, Jin L and Chen X (2020) The progress of gut microbiome research related to brain disorders, Journal of Neuroinflammation, 10.1186/s12974-020-1705-z, 17:1, Online publication date: 1-Dec-2020. Tan S, Chong C, Yap I, Thong K and Teh C (2020) Comparative assessment of faecal microbial composition and metabonome of swine, farmers and human control, Scientific Reports, 10.1038/s41598-020-65891-4, 10:1, Online publication date: 1-Dec-2020. He M, Tan C, Xu Y and Liu Y (2020) Gut microbiota-derived trimethylamine-N-oxide: A bridge between dietary fatty acid and cardiovascular disease?, Food Research International, 10.1016/j.foodres.2020.109812, 138, (109812), Online publication date: 1-Dec-2020. Herrema H and Niess J (2020) Intestinal microbial metabolites in human metabolism and type 2 diabetes, Diabetologia, 10.1007/s00125-020-05268-4, 63:12, (2533-2547), Online publication date: 1-Dec-2020. Andersson C, Liu C, Cheng S, Wang T, Gerszten R, Larson M and Vasan R (2020) Metabolomic signatures of cardiac remodelling and heart failure risk in the community, ESC Heart Failure, 10.1002/ehf2.12923, 7:6, (3707-3715), Online publication date: 1-Dec-2020. Fraser K, Roy N, Goumidi L, Verdu A, Suchon P, Leal-Valentim F, Trégouët D, Morange P and Martin J (2020) Plasma Biomarkers and Identification of Resilient Metabolic Disruptions in Patients With Venous Thromboembolism Using a Metabolic Systems Approach, Arteriosclerosis, Thrombosis, and Vascular Biology, 40:10, (2527-2538), Online publication date: 1-Oct-2020. Dannenberg L, Zikeli D, Benkhoff M, Ahlbrecht S, Kelm M, Levkau B and Polzin A (2020) Targeting the human microbiome and its metabolite TMAO in cardiovascular prevention and therapy, Pharmacology & Therapeutics, 10.1016/j.pharmthera.2020.107584, 213, (107584), Online publication date: 1-Sep-2020. Simó C and García-Cañas V (2020) Dietary bioactive ingredients to modulate the gut microbiota-derived metabolite TMAO. New opportunities for functional food development, Food & Function, 10.1039/D0FO01237H, 11:8, (6745-6776) Govindarajulu M, Pinky P, Steinke I, Bloemer J, Ramesh S, Kariharan T, Rella R, Bhattacharya S, Dhanasekaran M, Suppiramaniam V and Amin R (2020) Gut Metabolite TMAO Induces Synaptic Plasticity Deficits by Promoting Endoplasmic Reticulum Stress, Frontiers in Molecular Neuroscience, 10.3389/fnmol.2020.00138, 13 Witkowski M, Weeks T and Hazen S (2020) Gut Microbiota and Cardiovascular Disease, Circulation Research, 127:4, (553-570), Online publication date: 31-Jul-2020. Luz P, Haas E and Favarato D (2020)(2020) Intestinal Microbiota and Cardiovascular Diseases, International Journal of Cardiovascular Sciences, 10.36660/ijcs.20200043, Online publication date: 7-Jul-2020., Online publication date: 7-Jul-2020. Andraos S, Lange K, Clifford S, Jones B, Thorstensen E, Kerr J, Wake M, Saffery R, Burgner D and O'Sullivan J (2020) Plasma Trimethylamine N-Oxide and Its Precursors: Population Epidemiology, Parent–Child Concordance, and Associations with Reported Dietary Intake in 11- to 12-Year-Old Children and Their Parents, Current Developments in Nutrition, 10.1093/cdn/nzaa103, 4:7, Online publication date: 1-Jul-2020. Pathak P, Helsley R, Brown A, Buffa J, Choucair I, Nemet I, Gogonea C, Gogonea V, Wang Z, Garcia-Garcia J, Cai L, Temel R, Sangwan N, Hazen S and Brown J (2020) Small molecule inhibition of gut microbial choline trimethylamine lyase activity alters host cholesterol and bile acid metabolism, American Journal of Physiology-Heart and Circulatory Physiology, 10.1152/ajpheart.00584.2019, 318:6, (H1474-H1486), Online publication date: 1-Jun-2020. Liu C, Cheng L, Ji L, Li F, Zhan Y, Wu B, Ke Y, Chen P, Hua F, Yuan L, Min Z, Sun L, Chen H and Cheng Y (2020) Intestinal microbiota dysbiosis play a role in pathogenesis of patients with primary immune thrombocytopenia, Thrombosis Research, 10.1016/j.thromres.2020.03.012, 190, (11-19), Online publication date: 1-Jun-2020. Prame Kumar K and Wong C (2020) Imbalance in the force: the dark side of the microbiota on stroke risk and progression, Current Opinion in Neurobiology, 10.1016/j.conb.2019.10.002, 62, (10-16), Online publication date: 1-Jun-2020. Organ C, Li Z, Sharp T, Polhemus D, Gupta N, Goodchild T, Tang W, Hazen S and Lefer D (2020) Nonlethal Inhibition of Gut Microbial Trimethylamine N‐oxide Production Improves Cardiac Function and Remodeling in a Murine Model of Heart Failure, Journal of the American Heart Association, 9:10, Online publication date: 18-May-2020.Gencer B, Li X, Gurmu Y, Bonaca M, Morrow D, Cohen M
DOI: 10.1001/jamacardio.2017.2198
2017
Cited 204 times
Efficacy and Safety of Spironolactone in Acute Heart Failure
<h3>Importance</h3> Persistent congestion is associated with worse outcomes in acute heart failure (AHF). Mineralocorticoid receptor antagonists administered at high doses may relieve congestion, overcome diuretic resistance, and mitigate the effects of adverse neurohormonal activation in AHF. <h3>Objective</h3> To assess the effect of high-dose spironolactone and usual care on N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels compared with usual care alone. <h3>Design, Setting, and Participants</h3> This double-blind and placebo (or low-dose)-controlled randomized clinical trial was conducted in 22 US acute care hospitals among patients with AHF who were previously receiving no or low-dose (12.5 mg or 25 mg daily) spironolactone and had NT-proBNP levels of 1000 pg/mL or more or B-type natriuretic peptide levels of 250 pg/mL or more, regardless of ejection fraction. <h3>Interventions</h3> High-dose spironolactone (100 mg) vs placebo or 25 mg spironolactone (usual care) daily for 96 hours <h3>Main Outcomes and Measures</h3> The primary end point was the change in NT-proBNP levels from baseline to 96 hours. Secondary end points included the clinical congestion score, dyspnea assessment, net urine output, and net weight change. Safety end points included hyperkalemia and changes in renal function. <h3>Results</h3> A total of 360 patients were randomized, of whom the median age was 65 years, 129 (36%) were women, 200 (55.5%) were white, 151 (42%) were black, 8 (2%) were Hispanic or Latino, 9 (2.5%) were of other race/ethnicity, and the median left ventricular ejection fraction was 34%. Baseline median (interquartile range) NT-proBNP levels were 4601 (2697-9596) pg/mL among the group treated with high-dose spironolactone and 3753 (1968-7633) pg/mL among the group who received usual care. There was no significant difference in the log NT-proBNP reduction between the 2 groups (−0.55 [95% CI, −0.92 to −0.18] with high-dose spironolactone and −0.49 [95% CI, −0.98 to −0.14] with usual care,<i>P</i> = .57). None of the secondary end point or day-30 all-cause mortality or heart failure hospitalization rate differed between the 2 groups. The changes in serum potassium and estimated glomerular filtration rate at 24, 48, 72, and 96 hours. were similar between the 2 groups. <h3>Conclusions and Relevance</h3> Adding treatment with high-dose spironolactone to usual care for patients with AHF for 96 hours was well tolerated but did not improve the primary or secondary efficacy end points. <h3>Trial Registration</h3> clinicaltrials.gov Identifier:NCT02235077
DOI: 10.1161/jaha.115.002816
2016
Cited 201 times
Intestinal Microbiota‐Generated Metabolite Trimethylamine‐ <i>N‐</i> Oxide and 5‐Year Mortality Risk in Stable Coronary Artery Disease: The Contributory Role of Intestinal Microbiota in a COURAGE‐Like Patient Cohort
Background Trimethylamine‐ N ‐oxide ( TMAO ), a metabolite derived from gut microbes and dietary phosphatidylcholine, is linked to both coronary artery disease pathogenesis and increased cardiovascular risks. The ability of plasma TMAO to predict 5‐year mortality risk in patients with stable coronary artery disease has not been reported. This study examined the clinical prognostic value of TMAO in patients with stable coronary artery disease who met eligibility criteria for a patient cohort similar to that of the Clinical Outcomes Utilizing Revascularization and Aggressive Drug Evaluation (COURAGE) trial. Methods and Results We examined the relationship between fasting plasma TMAO and all‐cause mortality over 5‐year follow‐up in sequential patients with stable coronary artery disease (n=2235) who underwent elective coronary angiography. We identified the COURAGE ‐like patient cohort as patients who had evidence of significant coronary artery stenosis and who were managed with optimal medical treatment. Higher plasma TMAO levels were associated with a 4‐fold increased mortality risk. Following adjustments for traditional risk factors, high‐sensitivity C‐reactive protein, and estimated glomerular filtration rate, elevated TMAO levels remained predictive of 5‐year all‐cause mortality risk (quartile 4 versus 1, adjusted hazard ratio 1.95, 95% CI 1.33–2.86; P =0.003). TMAO remained predictive of incident mortality risk following cardiorenal and inflammatory biomarker adjustments to the model (adjusted hazard ratio 1.71, 95% CI 1.11–2.61; P =0.0138) and provided significant incremental prognostic value for all‐cause mortality (net reclassification index 42.37%, P &lt;0.001; improvement in area under receiver operator characteristic curve 70.6–73.76%, P &lt;0.001). Conclusions Elevated plasma TMAO levels portended higher long‐term mortality risk among patients with stable coronary artery disease managed with optimal medical treatment.
DOI: 10.1038/s41598-020-72685-1
2020
Cited 197 times
Machine learning prediction in cardiovascular diseases: a meta-analysis
Several machine learning (ML) algorithms have been increasingly utilized for cardiovascular disease prediction. We aim to assess and summarize the overall predictive ability of ML algorithms in cardiovascular diseases. A comprehensive search strategy was designed and executed within the MEDLINE, Embase, and Scopus databases from database inception through March 15, 2019. The primary outcome was a composite of the predictive ability of ML algorithms of coronary artery disease, heart failure, stroke, and cardiac arrhythmias. Of 344 total studies identified, 103 cohorts, with a total of 3,377,318 individuals, met our inclusion criteria. For the prediction of coronary artery disease, boosting algorithms had a pooled area under the curve (AUC) of 0.88 (95% CI 0.84-0.91), and custom-built algorithms had a pooled AUC of 0.93 (95% CI 0.85-0.97). For the prediction of stroke, support vector machine (SVM) algorithms had a pooled AUC of 0.92 (95% CI 0.81-0.97), boosting algorithms had a pooled AUC of 0.91 (95% CI 0.81-0.96), and convolutional neural network (CNN) algorithms had a pooled AUC of 0.90 (95% CI 0.83-0.95). Although inadequate studies for each algorithm for meta-analytic methodology for both heart failure and cardiac arrhythmias because the confidence intervals overlap between different methods, showing no difference, SVM may outperform other algorithms in these areas. The predictive ability of ML algorithms in cardiovascular diseases is promising, particularly SVM and boosting algorithms. However, there is heterogeneity among ML algorithms in terms of multiple parameters. This information may assist clinicians in how to interpret data and implement optimal algorithms for their dataset.
DOI: 10.1016/j.celrep.2017.05.077
2017
Cited 195 times
The TMAO-Producing Enzyme Flavin-Containing Monooxygenase 3 Regulates Obesity and the Beiging of White Adipose Tissue
<h2>Summary</h2> Emerging evidence suggests that microbes resident in the human intestine represent a key environmental factor contributing to obesity-associated disorders. Here, we demonstrate that the gut microbiota-initiated trimethylamine N-oxide (TMAO)-generating pathway is linked to obesity and energy metabolism. In multiple clinical cohorts, systemic levels of TMAO were observed to strongly associate with type 2 diabetes. In addition, circulating TMAO levels were associated with obesity traits in the different inbred strains represented in the Hybrid Mouse Diversity Panel. Further, antisense oligonucleotide-mediated knockdown or genetic deletion of the TMAO-producing enzyme flavin-containing monooxygenase 3 (FMO3) conferred protection against obesity in mice. Complimentary mouse and human studies indicate a negative regulatory role for FMO3 in the beiging of white adipose tissue. Collectively, our studies reveal a link between the TMAO-producing enzyme FMO3 and obesity and the beiging of white adipose tissue.
DOI: 10.1001/jama.2018.14852
2018
Cited 193 times
Effect of Inorganic Nitrite vs Placebo on Exercise Capacity Among Patients With Heart Failure With Preserved Ejection Fraction
There are few effective treatments for heart failure with preserved ejection fraction (HFpEF). Short-term administration of inorganic nitrite or nitrate preparations has been shown to enhance nitric oxide signaling, which may improve aerobic capacity in HFpEF.To determine the effect of 4 weeks' administration of inhaled, nebulized inorganic nitrite on exercise capacity in HFpEF.Multicenter, double-blind, placebo-controlled, 2-treatment, crossover trial of 105 patients with HFpEF. Participants were enrolled from July 22, 2016, to September 12, 2017, at 17 US sites, with final date of follow-up of January 2, 2018.Inorganic nitrite or placebo administered via micronebulizer device. During each 6-week phase of the crossover study, participants received no study drug for 2 weeks (baseline/washout) followed by study drug (nitrite or placebo) at 46 mg 3 times a day for 1 week followed by 80 mg 3 times a day for 3 weeks.The primary end point was peak oxygen consumption (mL/kg/min). Secondary end points included daily activity levels assessed by accelerometry, health status as assessed by the Kansas City Cardiomyopathy Questionnaire (score range, 0-100, with higher scores reflecting better quality of life), functional class, cardiac filling pressures assessed by echocardiography, N-terminal fragment of the prohormone brain natriuretic peptide levels, other exercise indices, adverse events, and tolerability. Outcomes were assessed after treatment for 4 weeks.Among 105 patients who were randomized (median age, 68 years; 56% women), 98 (93%) completed the trial. During the nitrite phase, there was no significant difference in mean peak oxygen consumption as compared with the placebo phase (13.5 vs 13.7 mL/kg/min; difference, -0.20 [95% CI, -0.56 to 0.16]; P = .27). There were no significant between-treatment phase differences in daily activity levels (5497 vs 5503 accelerometry units; difference, -15 [95% CI, -264 to 234]; P = .91), Kansas City Cardiomyopathy Questionnaire Clinical Summary Score (62.6 vs 61.9; difference, 1.1 [95% CI, -1.4 to 3.5]; P = .39), functional class (2.5 vs 2.5; difference, 0.1 [95% CI, -0.1 to 0.2]; P = .43), echocardiographic E/e' ratio (16.4 vs 16.6; difference, 0.1 [95% CI, -1.2 to 1.3]; P = .93), or N-terminal fragment of the prohormone brain natriuretic peptide levels (520 vs 533 pg/mL; difference, 11 [95% CI, -53 to 75]; P = .74). Worsening heart failure occurred in 3 participants (2.9%) during the nitrite phase and 8 (7.6%) during the placebo phase.Among patients with HFpEF, administration of inhaled inorganic nitrite for 4 weeks, compared with placebo, did not result in significant improvement in exercise capacity.ClinicalTrials.gov Identifier: NCT02742129.
DOI: 10.1016/j.jacc.2016.03.546
2016
Cited 185 times
Plasma Trimethylamine N -Oxide, a Gut Microbe–Generated Phosphatidylcholine Metabolite, Is Associated With Atherosclerotic Burden
Trimethylamine N-oxide (TMAO), a gut microbiota metabolite from dietary phosphatidylcholine, has mechanistic links to atherosclerotic coronary artery disease (CAD) pathogenesis and is associated with adverse outcomes. This study sought to examine the relationship between plasma TMAO levels and the complexity and burden of CAD and degree of subclinical myonecrosis. We studied 353 consecutive stable patients with evidence of atherosclerotic CAD detected by elective coronary angiography between 2012 and 2014. Their high-sensitivity cardiac troponin T (hs-cTnT) levels were measured. SYNTAX (Synergy Between Percutaneous Coronary Intervention With Taxus and Cardiac Surgery) scores and lesion characteristics were used to quantify atherosclerotic burden. Fasting plasma TMAO was measured by mass spectrometry. In this prospective cohort study, the median TMAO level was 5.5 μM (interquartile range [IQR]: 3.4 to 9.8 μM), the median SYNTAX score was 11.0 (IQR: 4.0 to 18.5), and 289 (81.9%), 40 (11.3%), and 24 (6.8%) patients had low (0 to 22), intermediate (23 to 32), and high (≥33) SYNTAX scores, respectively. Plasma TMAO levels correlated (all p < 0.0001) with the SYNTAX score (r = 0.61), SYNTAX score II (r = 0.62), and hs-cTnT (r = 0.29). Adjusting for traditional risk factors, body mass index, medications, lesion characteristic, renal function, and high-sensitivity C-reactive protein, elevated TMAO levels remained independently associated with a higher SYNTAX score (odds ratio [OR]: 4.82; p < 0.0001), SYNTAX score II (OR: 1.88; p = 0.0001), but were not associated with subclinical myonecrosis (OR: 1.14; p = 0.3147). Elevated TMAO level was an independent predictor of the presence of diffuse lesions, even after adjustments for traditional risk factors and for hs-cTnT (OR: 2.05; 95% confidence interval: 1.45 to 2.90; p = 0.0001). Fasting plasma TMAO levels are an independent predictor of a high atherosclerotic burden in patients with CAD.
DOI: 10.1373/clinchem.2016.263640
2017
Cited 183 times
Increased Trimethylamine N-Oxide Portends High Mortality Risk Independent of Glycemic Control in Patients with Type 2 Diabetes Mellitus
Abstract BACKGROUND Recent studies show a mechanistic link between intestinal microbial metabolism of dietary phosphatidylcholine and coronary artery disease pathogenesis. Concentrations of a proatherogenic gut microbe-generated metabolite, trimethylamine N-oxide (TMAO), predict increased incident cardiovascular disease risks in multiple cohorts. TMAO concentrations are increased in patients with type 2 diabetes mellitus (T2DM), but their prognostic value and relation to glycemic control are unclear. METHODS We examined the relationship between fasting TMAO and 2 of its nutrient precursors, choline and betaine, vs 3-year major adverse cardiac events and 5-year mortality in 1216 stable patients with T2DM who underwent elective diagnostic coronary angiography. RESULTS TMAO [4.4 μmol/L (interquartile range 2.8–7.7 μmol/L) vs 3.6 (2.3–5.7 μmol/L); P &amp;lt; 0.001] and choline concentrations were higher in individuals with T2DM vs healthy controls. Within T2DM patients, higher plasma TMAO was associated with a significant 3.0-fold increased 3-year major adverse cardiac event risk (P &amp;lt; 0.001) and a 3.6-fold increased 5-year mortality risk (P &amp;lt; 0.001). Following adjustments for traditional risk factors and high-sensitivity C-reactive protein, glycohemoglobin, and estimated glomerular filtration rate, increased TMAO concentrations remained predictive of both major adverse cardiac events and mortality risks in T2DM patients [e.g., quartiles 4 vs 1, hazard ratio 2.05 (95% CI, 1.31–3.20), P &amp;lt; 0.001; and 2.07 (95% CI, 1.37–3.14), P &amp;lt; 0.001, respectively]. CONCLUSIONS Fasting plasma concentrations of the proatherogenic gut microbe-generated metabolite TMAO are higher in diabetic patients and portend higher major adverse cardiac events and mortality risks independent of traditional risk factors, renal function, and relationship to glycemic control.
DOI: 10.1016/j.cardfail.2015.09.017
2015
Cited 180 times
Intersections Between Microbiome and Heart Failure: Revisiting the Gut Hypothesis
Microbes play an important role in human health and disease. In the setting of heart failure (HF), substantial hemodynamic changes, such as hypoperfusion and congestion in the intestines, can alter gut morphology, permeability, function, and possibly the growth and composition of gut microbiota. These changes can disrupt the barrier function of the intestines and exacerbate systemic inflammation via microbial or endotoxin translocation into systemic circulation. Furthermore, cardiorenal alterations via metabolites derived from gut microbiota can potentially mediate or modulate HF pathophysiology. Recently, trimethylamine N-oxide (TMAO) has emerged as a key mediator that provides a mechanistic link between gut microbiota and multiple cardiovascular diseases, including HF. Potential intervention strategies which may target this microbiota-driven pathology include dietary modification, prebiotics/probiotics, and selective binders of microbial enzymes or molecules, but further investigations into their safety and efficacy are warranted.
DOI: 10.1021/acs.analchem.8b05592
2019
Cited 165 times
Systematic Error Removal Using Random Forest for Normalizing Large-Scale Untargeted Lipidomics Data
Large-scale untargeted lipidomics experiments involve the measurement of hundreds to thousands of samples. Such data sets are usually acquired on one instrument over days or weeks of analysis time. Such extensive data acquisition processes introduce a variety of systematic errors, including batch differences, longitudinal drifts, or even instrument-to-instrument variation. Technical data variance can obscure the true biological signal and hinder biological discoveries. To combat this issue, we present a novel normalization approach based on using quality control pool samples (QC). This method is called systematic error removal using random forest (SERRF) for eliminating the unwanted systematic variations in large sample sets. We compared SERRF with 15 other commonly used normalization methods using six lipidomics data sets from three large cohort studies (832, 1162, and 2696 samples). SERRF reduced the average technical errors for these data sets to 5% relative standard deviation. We conclude that SERRF outperforms other existing methods and can significantly reduce the unwanted systematic variation, revealing biological variance of interest.
DOI: 10.1002/ejhf.1621
2019
Cited 163 times
Phenomapping of patients with heart failure with preserved ejection fraction using machine learning‐based unsupervised cluster analysis
Abstract Aim To identify distinct phenotypic subgroups in a highly‐dimensional, mixed‐data cohort of individuals with heart failure (HF) with preserved ejection fraction (HFpEF) using unsupervised clustering analysis. Methods and results The study included all Treatment of Preserved Cardiac Function Heart Failure with an Aldosterone Antagonist (TOPCAT) participants from the Americas ( n = 1767). In the subset of participants with available echocardiographic data (derivation cohort, n = 654), we characterized three mutually exclusive phenogroups of HFpEF participants using penalized finite mixture model‐based clustering analysis on 61 mixed‐data phenotypic variables. Phenogroup 1 had higher burden of co‐morbidities, natriuretic peptides, and abnormalities in left ventricular structure and function; phenogroup 2 had lower prevalence of cardiovascular and non‐cardiac co‐morbidities but higher burden of diastolic dysfunction; and phenogroup 3 had lower natriuretic peptide levels, intermediate co‐morbidity burden, and the most favourable diastolic function profile. In adjusted Cox models, participants in phenogroup 1 (vs. phenogroup 3) had significantly higher risk for all adverse clinical events including the primary composite endpoint, all‐cause mortality, and HF hospitalization. Phenogroup 2 (vs. phenogroup 3) was significantly associated with higher risk of HF hospitalization but a lower risk of atherosclerotic event (myocardial infarction, stroke, or cardiovascular death), and comparable risk of mortality. Similar patterns of association were also observed in the non‐echocardiographic TOPCAT cohort (internal validation cohort, n = 1113) and an external cohort of patients with HFpEF [Phosphodiesterase‐5 Inhibition to Improve Clinical Status and Exercise Capacity in Heart Failure with Preserved Ejection Fraction (RELAX) trial cohort, n = 198], with the highest risk of adverse outcome noted in phenogroup 1 participants. Conclusions Machine learning‐based cluster analysis can identify phenogroups of patients with HFpEF with distinct clinical characteristics and long‐term outcomes.
DOI: 10.1161/circulationaha.117.031560
2018
Cited 156 times
Prognostic Value of High-Sensitivity Troponin T in Chronic Heart Failure
Background: Most patients with chronic heart failure have detectable troponin concentrations when evaluated by high-sensitivity assays. The prognostic relevance of this finding has not been clearly established so far. We aimed to assess high-sensitivity troponin assay for risk stratification in chronic heart failure through a meta-analysis approach. Methods: Medline, EMBASE, Cochrane Library, and Scopus were searched in April 2017 by 2 independent authors. The terms were “troponin” AND “heart failure” OR “cardiac failure” OR “cardiac dysfunction” OR “cardiac insufficiency” OR “left ventricular dysfunction.” Inclusion criteria were English language, clinical stability, use of a high-sensitivity troponin assay, follow-up studies, and availability of individual patient data after request to authors. Data retrieved from articles and provided by authors were used in agreement with the PRISMA statement. The end points were all-cause death, cardiovascular death, and hospitalization for cardiovascular cause. Results: Ten studies were included, reporting data on 11 cohorts and 9289 patients (age 66±12 years, 77% men, 60% ischemic heart failure, 85% with left ventricular ejection fraction &lt;40%). High-sensitivity troponin T data were available for all patients, whereas only 209 patients also had high-sensitivity troponin I assayed. When added to a prognostic model including established risk markers (sex, age, ischemic versus nonischemic etiology, left ventricular ejection fraction, estimated glomerular filtration rate, and N-terminal fraction of pro-B-type natriuretic peptide), high-sensitivity troponin T remained independently associated with all-cause mortality (hazard ratio, 1.48; 95% confidence interval, 1.41–1.55), cardiovascular mortality (hazard ratio, 1.40; 95% confidence interval, 1.33–1.48), and cardiovascular hospitalization (hazard ratio, 1.42; 95% confidence interval, 1.36–1.49), over a median 2.4-year follow-up (all P &lt;0.001). High-sensitivity troponin T significantly improved risk prediction when added to a prognostic model including the variables above. It also displayed an independent prognostic value for all outcomes in almost all population subgroups. The area under the curve–derived 18 ng/L cutoff yielded independent prognostic value for the 3 end points in both men and women, patients with either ischemic or nonischemic etiology, and across categories of renal dysfunction. Conclusions: In chronic heart failure, high-sensitivity troponin T is a strong and independent predictor of all-cause and cardiovascular mortality, and of hospitalization for cardiovascular causes, as well. This biomarker then represents an additional tool for prognostic stratification.
DOI: 10.1016/j.cardfail.2016.06.423
2016
Cited 142 times
Relevance of Changes in Serum Creatinine During a Heart Failure Trial of Decongestive Strategies: Insights From the DOSE Trial
Worsening renal function (WRF) is a common endpoint in decompensated heart failure clinical trials because of associations between WRF and adverse outcomes. However, WRF has not universally been identified as a poor prognostic sign, challenging the validity of WRF as a surrogate endpoint. Our aim was to describe the associations between changes in creatinine and adverse outcomes in a clinical trial of decongestive therapies.We investigated the association between changes in creatinine and the composite endpoint of death, rehospitalization or emergency room visit within 60 days in 301 patients in the Diuretic Optimization Strategies Evaluation (DOSE) trial. WRF was defined as an increase in creatinine >0.3 mg/dL and improvement in renal function (IRF) as a decrease >0.3 mg/dL. When examining linear changes in creatinine from baseline to 72 hours (the coprimary endpoint of DOSE), increasing creatinine was associated with lower risk for the composite outcome (HR = 0.81 per 0.3 mg/dL increase, 95% CI 0.67-0.98, P = .026). Compared with patients with stable renal function (n = 219), WRF (n = 54) was not associated with the composite endpoint (HR = 1.17, 95% CI = 0.77-1.78, P = .47). However, compared with stable renal function, there was a strong relationship between IRF (n = 28) and the composite endpoint (HR = 2.52, 95% CI = 1.57-4.03, P < .001).The coprimary endpoint of the DOSE trial, a linear increase in creatinine, was paradoxically associated with improved outcomes. This was driven by absence of risk attributable to WRF and a strong risk associated with IRF. These results argue against using changes in serum creatinine as a surrogate endpoint in trials of decongestive strategies.
DOI: 10.1097/hco.0000000000000445
2017
Cited 141 times
Gut microbiome and its role in cardiovascular diseases
In recent years, an interest in intestinal microbiota-host interactions has increased due to many findings about the impact of gut bacteria on human health and disease. Dysbiosis, a change in the composition of the gut microbiota, has been associated with much pathology, including cardiovascular diseases (CVD). This article will review normal functions of the gut microbiome, its link to CVD, and potential therapeutic interventions.The recently discovered contribution of gut microbiota-derived molecules in the development of heart disease and its risk factors has significantly increased attention towards the connection between our gut and heart. The gut microbiome is virtually an endocrine organ, arguably the largest, capable of contributing to and reacting to circulating signaling molecules within the host. Gut microbiota-host interactions occur through many pathways, including trimethylamine-N-oxide and short-chain fatty acids. These molecules and others have been linked to much pathology including chronic kidney disease, atherosclerosis, and hypertension.Although our understanding of gut microbiota-host interactions has increased recently; many questions remain about the mechanistic links between the gut microbiome and CVD. With further research, we may one day be able to add gut microbiota profiles as an assessable risk factor for CVD and target therapies towards the gut microbiota.
DOI: 10.1172/jci.insight.99096
2018
Cited 127 times
Untargeted metabolomics identifies trimethyllysine, a TMAO-producing nutrient precursor, as a predictor of incident cardiovascular disease risk
Using an untargeted metabolomics approach in initial (N = 99 subjects) and replication cohorts (N = 1,162), we discovered and structurally identified a plasma metabolite associated with cardiovascular disease (CVD) risks, N6,N6,N6-trimethyl-L-lysine (trimethyllysine, TML). Stable-isotope-dilution tandem mass spectrometry analyses of an independent validation cohort (N = 2,140) confirmed TML levels are independently associated with incident (3-year) major adverse cardiovascular event risks (hazards ratio [HR], 2.4; 95% CI, 1.7–3.4) and incident (5-year) mortality risk (HR, 2.9; 95% CI, 2.0–4.2). Genome-wide association studies identified several suggestive loci for TML levels, but none reached genome-wide significance; and d9(trimethyl)-TML isotope tracer studies confirmed TML can serve as a nutrient precursor for gut microbiota–dependent generation of trimethylamine (TMA) and the atherogenic metabolite trimethylamine N-oxide (TMAO). Although TML was shown to be abundant in both plant- and animal-derived foods, mouse and human fecal cultures (omnivores and vegans) showed slow conversion of TML to TMA. Furthermore, unlike chronic dietary choline, TML supplementation in mice failed to elevate plasma TMAO or heighten thrombosis potential in vivo. Thus, TML is identified as a strong predictor of incident CVD risks in subjects and to serve as a dietary precursor for gut microbiota–dependent generation of TMAO; however, TML does not appear to be a major microbial source for TMAO generation in vivo.
DOI: 10.1161/atvbaha.120.314139
2020
Cited 105 times
Targeted Inhibition of Gut Microbial Trimethylamine N-Oxide Production Reduces Renal Tubulointerstitial Fibrosis and Functional Impairment in a Murine Model of Chronic Kidney Disease
Objective: Gut microbial metabolism of dietary choline, a nutrient abundant in a Western diet, produces trimethylamine (TMA) and the atherothrombosis- and fibrosis-promoting metabolite TMA-N-oxide (TMAO). Recent clinical and animal studies reveal that elevated TMAO levels are associated with heightened risks for both cardiovascular disease and incident chronic kidney disease development. Despite this, studies focusing on therapeutically targeting gut microbiota-dependent TMAO production and its impact on preserving renal function are limited. Approach and Results: Herein we examined the impact of pharmacological inhibition of choline diet-induced gut microbiota-dependent production of TMA, and consequently TMAO, on renal tubulointerstitial fibrosis and functional impairment in a model of chronic kidney disease. Initial studies with a gut microbial choline TMA-lyase mechanism-based inhibitor, iodomethylcholine, confirmed both marked suppression of TMA generation, and consequently TMAO levels, and selective targeting of the gut microbial compartment (ie, both accumulation of the drug in intestinal microbes and limited systemic exposure in the host). Dietary supplementation of either choline or TMAO significantly augmented multiple indices of renal functional impairment and fibrosis associated with chronic subcutaneous infusion of isoproterenol. However, the presence of the gut microbiota-targeting inhibitor iodomethylcholine blocked choline diet-induced elevation in TMAO, and both significantly improved decline in renal function, and significantly attenuated multiple indices of tubulointerstitial fibrosis. Iodomethylcholine treatment also reversed many choline diet-induced changes in cecal microbial community composition associated with TMAO and renal functional impairment. Conclusions: Selective targeting of gut microbiota-dependent TMAO generation may prevent adverse renal structural and functional alterations in subjects at risk for chronic kidney disease.
DOI: 10.1016/j.hrthm.2023.03.1538
2023
Cited 89 times
2023 HRS/APHRS/LAHRS guideline on cardiac physiologic pacing for the avoidance and mitigation of heart failure
Cardiac physiologic pacing (CPP), encompassing cardiac resynchronization therapy (CRT) and conduction system pacing (CSP), has emerged as a pacing therapy strategy that may mitigate or prevent the development of heart failure (HF) in patients with ventricular dyssynchrony or pacing-induced cardiomyopathy. This clinical practice guideline is intended to provide guidance on indications for CRT for HF therapy and CPP in patients with pacemaker indications or HF, patient selection, pre-procedure evaluation and preparation, implant procedure management, follow-up evaluation and optimization of CPP response, and use in pediatric populations. Gaps in knowledge, pointing to new directions for future research, are also identified.
DOI: 10.1038/s41591-023-02223-9
2023
Cited 50 times
The artificial sweetener erythritol and cardiovascular event risk
DOI: 10.1161/circheartfailure.122.009972
2023
Cited 39 times
Gut Microbiota-Generated Phenylacetylglutamine and Heart Failure
Background: The gut microbiota-dependent metabolite phenylacetylgutamine (PAGln) is both associated with atherothrombotic heart disease in humans, and mechanistically linked to cardiovascular disease pathogenesis in animal models via modulation of adrenergic receptor signaling. Methods: Here we examined both clinical and mechanistic relationships between PAGln and heart failure (HF). First, we examined associations among plasma levels of PAGln and HF, left ventricular ejection fraction, and N-terminal pro-B-type natriuretic peptide in 2 independent clinical cohorts of subjects undergoing coronary angiography in tertiary referral centers (an initial discovery US Cohort, n=3256; and a validation European Cohort, n=829). Then, the impact of PAGln on cardiovascular phenotypes relevant to HF in cultured cardiomyoblasts, and in vivo were also examined. Results: Circulating PAGln levels were dose-dependently associated with HF presence and indices of severity (reduced ventricular ejection fraction, elevated N-terminal pro-B-type natriuretic peptide) independent of traditional risk factors and renal function in both cohorts. Beyond these clinical associations, mechanistic studies showed both PAGln and its murine counterpart, phenylacetylglycine, directly fostered HF-relevant phenotypes, including decreased cardiomyocyte sarcomere contraction, and B-type natriuretic peptide gene expression in both cultured cardiomyoblasts and murine atrial tissue. Conclusions: The present study reveals the gut microbial metabolite PAGln is clinically and mechanistically linked to HF presence and severity. Modulating the gut microbiome, in general, and PAGln production, in particular, may represent a potential therapeutic target for modulating HF. Registration: URL: https://clinicaltrials.gov/ ; Unique identifier: NCT00590200 and URL: https://drks.de/drks_web/ ; Unique identifier: DRKS00020915.
DOI: 10.1161/circulationaha.122.060573
2023
Cited 18 times
Gut Microbiota–Derived Trimethylamine N-Oxide Contributes to Abdominal Aortic Aneurysm Through Inflammatory and Apoptotic Mechanisms
Large-scale human and mechanistic mouse studies indicate a strong relationship between the microbiome-dependent metabolite trimethylamine N-oxide (TMAO) and several cardiometabolic diseases. This study aims to investigate the role of TMAO in the pathogenesis of abdominal aortic aneurysm (AAA) and target its parent microbes as a potential pharmacological intervention.TMAO and choline metabolites were examined in plasma samples, with associated clinical data, from 2 independent patient cohorts (N=2129 total). Mice were fed a high-choline diet and underwent 2 murine AAA models, angiotensin II infusion in low-density lipoprotein receptor-deficient (Ldlr-/-) mice or topical porcine pancreatic elastase in C57BL/6J mice. Gut microbial production of TMAO was inhibited through broad-spectrum antibiotics, targeted inhibition of the gut microbial choline TMA lyase (CutC/D) with fluoromethylcholine, or the use of mice genetically deficient in flavin monooxygenase 3 (Fmo3-/-). Finally, RNA sequencing of in vitro human vascular smooth muscle cells and in vivo mouse aortas was used to investigate how TMAO affects AAA.Elevated TMAO was associated with increased AAA incidence and growth in both patient cohorts studied. Dietary choline supplementation augmented plasma TMAO and aortic diameter in both mouse models of AAA, which was suppressed with poorly absorbed oral broad-spectrum antibiotics. Treatment with fluoromethylcholine ablated TMAO production, attenuated choline-augmented aneurysm initiation, and halted progression of an established aneurysm model. In addition, Fmo3-/- mice had reduced plasma TMAO and aortic diameters and were protected from AAA rupture compared with wild-type mice. RNA sequencing and functional analyses revealed choline supplementation in mice or TMAO treatment of human vascular smooth muscle cells-augmented gene pathways associated with the endoplasmic reticulum stress response, specifically the endoplasmic reticulum stress kinase PERK.These results define a role for gut microbiota-generated TMAO in AAA formation through upregulation of endoplasmic reticulum stress-related pathways in the aortic wall. In addition, inhibition of microbiome-derived TMAO may serve as a novel therapeutic approach for AAA treatment where none currently exist.
DOI: 10.1093/eurheartj/ehad333
2023
Cited 17 times
Atlas of gut microbe-derived products from aromatic amino acids and risk of cardiovascular morbidity and mortality
Abstract Aims Precision microbiome modulation as a novel treatment strategy is a rapidly evolving and sought goal. The aim of this study is to determine relationships among systemic gut microbial metabolite levels and incident cardiovascular disease risks to identify gut microbial pathways as possible targets for personalized therapeutic interventions. Methods and results Stable isotope dilution mass spectrometry methods to quantitatively measure aromatic amino acids and their metabolites were used to examine sequential subjects undergoing elective diagnostic cardiac evaluation in two independent cohorts with longitudinal outcome data [US (n = 4000) and EU (n = 833) cohorts]. It was also used in plasma from humans and mice before vs. after a cocktail of poorly absorbed antibiotics to suppress gut microbiota. Multiple aromatic amino acid-derived metabolites that originate, at least in part, from gut bacteria are associated with incident (3-year) major adverse cardiovascular event (MACE) risks (myocardial infarction, stroke, or death) and all-cause mortality independent of traditional risk factors. Key gut microbiota-derived metabolites associated with incident MACE and poorer survival risks include: (i) phenylacetyl glutamine and phenylacetyl glycine (from phenylalanine); (ii) p-cresol (from tyrosine) yielding p-cresol sulfate and p-cresol glucuronide; (iii) 4-OH-phenyllactic acid (from tyrosine) yielding 4-OH-benzoic acid and 4-OH-hippuric acid; (iv) indole (from tryptophan) yielding indole glucuronide and indoxyl sulfate; (v) indole-3-pyruvic acid (from tryptophan) yielding indole-3-lactic acid and indole-3-acetyl-glutamine, and (vi) 5-OH-indole-3-acetic acid (from tryptophan). Conclusion Key gut microbiota-generated metabolites derived from aromatic amino acids independently associated with incident adverse cardiovascular outcomes are identified, and thus will help focus future studies on gut-microbial metabolic outputs relevant to host cardiovascular health.
DOI: 10.1002/ejhf.3111
2024
Cited 3 times
Prognostic Value of Gut <scp>Microbe‐Generated</scp> Metabolite Phenylacetylglutamine in Patients with Heart Failure
Phenylacetylglutamine (PAGln) is a phenylalanine-derived metabolite produced by gut microbiota with mechanistic links to heart failure (HF)-relevant phenotypes. We sought to investigate the prognostic value of PAGln in patients with stable HF.Fasting plasma PAGln levels were measured by stable-isotope-dilution liquid chromatography-tandem mass spectrometry (LC-MS/MS) in patients with stable HF from two large cohorts. All-cause mortality was assessed at 5-year follow-up in the Cleveland cohort, and HF, hospitalization, or mortality were assessed at 3-year follow-up in the Berlin cohort. Within the Cleveland cohort, median PAGln levels were 4.2 (interquartile range [IQR] 2.4-6.9) μM. Highest quartile of PAGln was associated with 3.09-fold increased mortality risk compared to lowest quartile. Following adjustments for traditional risk factors, as well as race, estimated glomerular filtration rate, amino-terminal pro-B-type natriuretic peptide, high-sensitivity C-reactive protein, left ventricular ejection fraction, ischaemic aetiology, and HF drug treatment, elevated PAGln levels remained predictive of 5-year mortality in quartile comparisons (adjusted hazard ratio [HR] [95% confidence interval, CI] for Q4 vs Q1: 1.64 [1.07-2.53]). In the Berlin cohort, a similar distribution of PAGln levels was observed (median 3.2 [IQR 2.0-4.8] μM), and PAGln levels were associated with a 1.92-fold increase in 3-year HF hospitalization or all-cause mortality risk (adjusted HR [95% CI] for Q4 vs Q1: 1.92 [1.02-3.61]). Prognostic value of PAGln appears to be independent of trimethylamine N-oxide levels.High levels of PAGln are associated with adverse outcomes independent of traditional cardiac risk factors and cardio-renal risk markers.
DOI: 10.1161/01.cir.0000106903.98196.b6
2003
Cited 228 times
Plasma B-Type Natriuretic Peptide Levels in Ambulatory Patients With Established Chronic Symptomatic Systolic Heart Failure
The diagnostic and prognostic values of plasma B-type natriuretic peptide (BNP) testing are established. However, the range of plasma BNP levels present in the setting of chronic, stable systolic heart failure (HF) is unclear.We followed up 558 consecutive ambulatory patients with chronic, stable systolic HF (left ventricular ejection fraction <50%) treated at a specialized outpatient HF clinic between November 2001 and February 2003. Retrospective chart review was performed to determine clinical and functional data at the time of BNP testing (Biosite Triage). The clinical characteristics of patients with plasma BNP levels <100 pg/mL and those with > or =100 pg/mL were compared. In our cohort, 60 patients were considered asymptomatic, and their plasma BNP levels ranged from 5 to 572 pg/mL (median, 147 pg/mL). Of the remaining 498 symptomatic (NYHA functional class II-III) patients, 106 (21.3%) had plasma BNP levels in the "normal" diagnostic range (<100 pg/mL). Patients in this "normal BNP" subgroup were more likely to be younger, to be female, to have nonischemic pathogenesis, and to have better-preserved cardiac and renal function and less likely to have atrial fibrillation.In the ambulatory care setting, both symptomatic and asymptomatic patients with chronic, stable systolic HF may present with a wide range of plasma BNP levels. In a subset of symptomatic patients (up to 21% in our cohort), plasma BNP levels are below what would be considered "diagnostic" (<100 pg/mL).
DOI: 10.1373/clinchem.2007.095679
2007
Cited 218 times
National Academy of Clinical Biochemistry Laboratory Medicine Practice Guidelines: Use of Cardiac Troponin and B-Type Natriuretic Peptide or N-Terminal proB-Type Natriuretic Peptide for Etiologies Other than Acute Coronary Syndromes and Heart Failure
Over the past decade, cardiac troponin (cTn)1 has become the cornerstone laboratory medicine measurement for assessment of myocardial infarction (MI) in suspected acute coronary syndrome (ACS) patients. In the past 5–7 years, methods for measuring the natriuretic peptide B-type natriuretic peptide (BNP) and its inert cometabolite N-terminal proBNP (NT-proBNP) have become available, and much knowledge has accumulated regarding their clinical use in the context of heart failure and hemodynamic stress. In addition to ACS and heart failure, there are common and clinically important patient cohorts in whom these measurements can aid in diagnosis and management. For this reason, the National Academy of Clinical Biochemistry (NACB) formed a Laboratory Medicine Practice Guidelines (LMPG) committee to extend cardiac biochemical marker recommendations and establish modern guidelines for utilization …
DOI: 10.1016/j.cardfail.2009.07.003
2010
Cited 207 times
Serum Neutrophil Gelatinase-Associated Lipocalin (NGAL) in Predicting Worsening Renal Function in Acute Decompensated Heart Failure
The development of worsening renal function (WRF, defined as creatinine rise >or=0.3mg/dL) occurs frequently in the setting of acute decompensated heart failure (ADHF) and strongly predicts adverse clinical outcomes. Neutrophil gelatinase-associated lipocalin (NGAL) is produced by the nephron in response to tubular epithelial damage and serves as an early marker for acute renal tubular injury. We sought to determine the relationship between admission serum NGAL levels and WRF in the setting of ADHF.We measured serum NGAL levels in 91 patients admitted to the hospital with ADHF. Patients were adjudicated by independent physician into those that did or did not develop WRF over the ensuing 5 days of in-hospital treatment. In our study cohort (68% male, mean age 61+/-15 years, mean left ventricular ejection fraction 31+/-14%), median admission serum NGAL level was 165 ng/mL (interquartile range [IQR] 108-235 ng/mL). Thirty-five patients (38%) developed WRF within the 5-day follow-up. Patients who developed WRF versus those without WRF had significantly higher median admission serum NGAL levels (194 [IQR 150-292] ng/mL vs. 128 [IQR 97-214] ng/mL, P=.001). High serum NGAL levels at admission were associated with greater likelihood of developing WRF (odds ratio: 1.92, 95% confidence interval 1.23-3.12, P=.004). In particular, admission NGAL >or=140 ng/mL had a 7.4-fold increase in risk of developing WRF, with a sensitivity and specificity of 86% and 54%, respectively.The presence of elevated admission serum NGAL levels is associated with heightened risk of subsequent development of WRF in patients admitted with ADHF.
DOI: 10.1016/j.jacc.2008.02.083
2008
Cited 191 times
Sodium Nitroprusside for Advanced Low-Output Heart Failure
This study was designed to examine the safety and efficacy of sodium nitroprusside (SNP) for patients with acute decompensated heart failure (ADHF) and low-output states. Inotropic therapy has been predominantly used in the management of patients with ADHF presenting with low cardiac output. We reviewed all consecutive patients with ADHF admitted between 2000 and 2005 with a cardiac index ≤2 l/min/m2 for intensive medical therapy including vasoactive drugs. Administration of SNP was chosen by the attending clinician, nonrandomized, and titrated to a target mean arterial pressure of 65 to 70 mm Hg. Compared with control patients (n = 97), cases treated with SNP (n = 78) had significantly higher mean central venous pressure (15 vs. 13 mm Hg; p = 0.001), pulmonary capillary wedge pressure (29 vs. 24 mm Hg; p = 0.001), but similar demographics, medications, and renal function at baseline. Use of SNP was not associated with higher rates of inotropic support or worsening renal function during hospitalization. Patients treated with SNP achieved greater improvement in hemodynamic measurements during hospitalization, had higher rates of oral vasodilator prescription at discharge, and had lower rates of all-cause mortality (29% vs. 44%; odds ratio: 0.48; p = 0.005; 95% confidence interval: 0.29 to 0.80) without increase in rehospitalization rates (58% vs. 56%; p = NS). In patients with advanced, low-output heart failure, vasodilator therapy used in conjunction with optimal current medical therapy during hospitalization might be associated with favorable long-term clinical outcomes irrespective of inotropic support or renal dysfunction and remains an excellent therapeutic choice in hospitalized ADHF patients.
DOI: 10.1016/j.cardfail.2009.01.014
2009
Cited 180 times
Soluble Angiotensin-Converting Enzyme 2 in Human Heart Failure: Relation With Myocardial Function and Clinical Outcomes
Angiotensin-converting enzyme 2 (ACE2) is an endogenous counterregulator of the renin-angiotensin system. The relationship between soluble ACE2 (sACE2), myocardial function, and clinical outcomes in patients with chronic systolic heart failure is not well established.We measured sACE2 activity in 113 patients with chronic systolic heart failure (left ventricular ejection fraction [LVEF] <or=35%, New York Heart Association Class II-IV). Comprehensive echocardiography was performed at the time of blood sampling. We prospectively examined adverse clinical events (death, cardiac transplant, and heart failure hospitalizations) over 34 +/- 17 months. Patients who had higher sACE2 plasma activity were more likely to have a lower LVEF (Spearman's r = -0.36, P < .001), greater right ventricular systolic dysfunction (r = 0.33, P < .001), higher estimated pulmonary artery systolic pressure (r = 0.35, P = .002), larger left ventricular end-diastolic diameter (r = 0.23, P = .02), and higher plasma NT-proBNP levels (r = 0.35, P < .001). sACE2 was less associated with diastolic dysfunction (r = 0.19, P = .05), and was similar between patients with ischemic and nonischemic cardiomyopathies. There was no relationship between sACE2 activity and markers of systemic inflammation. After adjusting for NT-proBNP and LVEF, sACE2 activity remained an independent predictor of adverse clinical events (HR = 1.7 [95% CI: 1.1-2.6], P = .018).Elevated plasma sACE2 activity was associated with greater severity of myocardial dysfunction and was an independent predictor of adverse clinical events.
DOI: 10.1016/j.jacc.2009.02.036
2009
Cited 176 times
Diminished Global Arginine Bioavailability and Increased Arginine Catabolism as Metabolic Profile of Increased Cardiovascular Risk
We hypothesized that an integrated assessment of arginine with its catabolic products might better predict cardiovascular risks than arginine levels alone. Arginine is the sole nitrogen source for nitric oxide (NO) synthesis. The major catabolic products of arginine are ornithine and citrulline. Plasma levels of free arginine, ornithine, citrulline, and the endogenous NO synthase inhibitor asymmetric dimethylarginine (ADMA) were measured with liquid chromatography coupled with tandem mass spectrometry. We examined the relationship of global arginine bioavailability ratio (GABR) (defined as arginine/[ornithine + citrulline]) versus arginine and its catabolic metabolites to prevalence of significantly obstructive coronary artery disease (CAD) and incidence of major adverse cardiovascular events (MACE) (death, myocardial infarction, stroke) over a 3-year follow-up in 1,010 subjects undergoing elective cardiac catheterization. Patients with significantly obstructive CAD had significantly lower GABR (median [interquartile range]: 1.06 [0.75 to 1.31] vs. 1.27 [0.96 to 1.73], p < 0.001) and arginine levels [mean: 68 ± 20 μmol/l vs. 74 ± 24 μmol/l, p < 0.001) than those without significantly obstructive CAD. After adjusting for Framingham risk score, C-reactive protein, and renal function, lower GABR (but not arginine levels) and higher citrulline levels remained significantly associated with both the prevalence of significantly obstructive CAD (adjusted odds ratio: 3.93, p < 0.001, and 5.98, p < 0.001, respectively) and 3-year risk for the incidence of MACE (adjusted hazard ratio: 1.98, p = 0.025, and 2.40, p = 0.01, respectively) and remained significant after adjusting for ADMA. GABR might serve as a more comprehensive concept of reduced NO synthetic capacity compared with systemic arginine levels. Diminished GABR and high citrulline levels are associated with both development of significantly obstructive atherosclerotic CAD and heightened long-term risk for MACE.
DOI: 10.1373/clinchem.2006.084715
2007
Cited 175 times
National Academy of Clinical Biochemistry and IFCC Committee for Standardization of Markers of Cardiac Damage Laboratory Medicine Practice Guidelines: Analytical Issues for Biochemical Markers of Acute Coronary Syndromes
### a. Background In 1999, the National Academy of Clinical Biochemistry (NACB)1 published the first standards of laboratory practice addressing analytical and clinical recommendations for use of cardiac markers in coronary artery diseases (1). The objectives were to recommend the appropriate implementation and utilization of cardiac biomarkers, specifically for cardiac troponin (cTn), which had just gained US Food and Drug Administration (FDA) clearance as a cardiac biomarker to aid in the diagnosis of acute myocardial infarction (AMI). In 2001, the IFCC Committee on Standardization of Markers of Cardiac Damage (C-SMCD) recommended quality specifications for analytical and preanalytical factors for cTn assays (2). The objectives were intended for use by the manufacturers of commercial assays and by clinical laboratories that use cTn assays. The overall goal was to establish uniform criteria so that all cTn assays could objectively be evaluated for their analytical qualities and clinical performance. These general principles can also be applied to creatine kinase MB (CK-MB) mass and myoglobin assays by use of the analytical recommendations in this document. In this report, we provide the background for establishing updated practice guidelines with recommendations addressing analytical issues for cardiac biomarkers based on 8 years of evidence-based medical and scientific observations since the publication of the initial recommendations (1). ### recommendations: analytical aspects of acs biomarkers ALL CLASS I 1. Reference decision-limits should be established for each cardiac biomarker based on a population of normal, healthy individuals without a known history of heart disease (reference population). For cardiac troponin …
DOI: 10.1016/j.amjcard.2006.04.018
2006
Cited 171 times
Plasma Myeloperoxidase Levels in Patients With Chronic Heart Failure
Increased oxidative stress and endothelial dysfunction are commonly observed in patients with chronic heart failure (HF). The relation between myeloperoxidase (MPO), an inflammatory marker with mechanistic links to plaque vulnerability and abnormal ventricular remodeling, and degrees of severity in chronic HF has not been reported. Plasma MPO levels were measured in 105 normal controls (no history of HF or left ventricular dysfunction) and 102 patients with chronic systolic HF (left ventricular ejection fraction <50%), and the relations among plasma MPO levels, plasma B-type natriuretic peptide levels, and the left ventricular ejection fraction were examined. Plasma MPO levels in patients with chronic systolic HF were significantly elevated compared with those of healthy controls (1,158 +/- 2,965 vs 204 +/- 139 pM, p <0.0001). Plasma MPO levels increased in parallel with increasing New York Heart Association class (p <0.0001) and were correlated with plasma B-type natriuretic peptide levels (Spearman's r = 0.39, p <0.0001). Levels of MPO were strongly associated with the prevalence of HF (unadjusted odds ratio 30.3, 95% confidence interval 11.1 to 94.5) and remained significant when adjusted for age and B-type natriuretic peptide (odds ratio 27.7, 95% confidence interval 3.6 to 371.1). In conclusion, in a cohort of patients with chronic HF, plasma MPO levels were elevated compared with those of normal controls and were associated with worsening functional class.
DOI: 10.1016/j.amjcard.2011.03.056
2011
Cited 169 times
Usefulness of Plasma Galectin-3 Levels in Systolic Heart Failure to Predict Renal Insufficiency and Survival
Galectin-3 plays an important role in fibroblast activation and fibrosis in animal models. Increased galectin-3 levels are associated with poor long-term survival in heart failure (HF). We examined the relation between plasma galectin-3 levels and myocardial indexes of systolic HF. We measured plasma galectin-3 in 133 subjects with chronic HF and 45 with advanced decompensated HF using echocardiographic and hemodynamic evaluations. In the chronic HF cohort, median plasma galectin-3 level was 13.9 ng/ml (interquartile range 12.1 to 16.9). Higher galectin-3 was associated with more advanced age (r = 0.22, p = 0.010), poor renal function (estimated glomerular filtration rate, r = -0.24, p = 0.007; cystatin C, r = 0.38, p <0.0001) and predicted all-cause mortality (hazard ratio 1.86, 95% confidence interval 1.36 to 2.54, p <0.001). In multivariate analysis, galectin-3 remained an independent predictor of all-cause mortality after adjusting for age, estimated glomerular filtration rate, left ventricular (LV) ejection fraction, and mitral early diastolic myocardial relaxation velocity at septal mitral annulus (hazard ratio 1.94, 95% confidence interval 1.30 to 2.91, p = 0.001). However, galectin-3 did not predict the combined end point of all-cause mortality, cardiac transplantation, or HF hospitalization (p >0.05). Furthermore, there were no relations between galectin-3 and LV end-diastolic volume index (r = -0.05, p = 0.61), LV ejection fraction (r = 0.10, p = 0.25), or LV diastolic function (mitral early diastolic myocardial relaxation velocity at septal mitral annulus, r = 0.06, p = 0.52; left atrial volume index, r = 0.08, p = 0.41). In the advanced decompensated HF cohort, we did not observe any relation between galectin-3 and echocardiographic or hemodynamic indexes. In conclusion, high plasma galectin-3 levels were associated with renal insufficiency and poorer survival in patients with chronic systolic HF. However, we did not observe a relation between galectin-3 and echocardiographic or hemodynamic indexes.
DOI: 10.1016/j.jacc.2007.02.053
2007
Cited 167 times
Prognostic Value and Echocardiographic Determinants of Plasma Myeloperoxidase Levels in Chronic Heart Failure
The purpose of this study was to explore the relationship between myeloperoxidase (MPO) and cardiac structure, performance, and prognosis. Myeloperoxidase is an inflammatory marker that is elevated in patients with heart failure (HF) and cardiac dysfunction, with mechanistic links to plaque vulnerability and left ventricular (LV) remodeling. We evaluated plasma MPO levels (CardioMPO, PrognostiX, Inc., Cleveland, Ohio) in 140 patients with chronic systolic HF (LV ejection fraction <35%) and examined the plasma MPO levels’ relationships with echocardiographic indexes of systolic and diastolic performance, as well as long-term clinical outcomes (death, cardiac transplantation, or HF hospitalization). Within the overall cohort, increasing plasma MPO levels were associated with increasing likelihood of more advanced HF (restrictive diastolic stage, right ventricular systolic dysfunction ≥3+, and tricuspid regurgitation area ≥1.8 cm2). Plasma MPO levels were predictive of long-term clinical outcomes (risk ratio [95% confidence interval] = 3.35 [1.52 to 8.86]), even after adjustment for age, LV ejection fraction, plasma B-type natriuretic peptide (BNP), creatinine clearance, or diastolic stage. In receiver-operator characteristic curve analyses, addition of MPO to BNP testing augmented the predictive accuracy of future adverse clinical events (area under the curve 0.66 for BNP only [chi-square test = 12.9, p = 0.0003], and 0.70 for BNP plus MPO [chi-square test = 15.87, p = 0.0004]). In chronic systolic HF, elevated plasma MPO levels are associated with an increased likelihood of more advanced HF. Moreover, elevated plasma MPO levels within a HF subject seem to be predictive of increased adverse clinical outcomes.
DOI: 10.1016/j.cardfail.2008.02.010
2008
Cited 159 times
Prompt Reduction in Intra-Abdominal Pressure Following Large-Volume Mechanical Fluid Removal Improves Renal Insufficiency in Refractory Decompensated Heart Failure
Our group recently reported that elevated intra-abdominal pressure (IAP, defined as > or = 8 mm Hg) can be associated with renal dysfunction in patients with advanced decompensated heart failure (ADHF). We hypothesize that in the setting of persistently elevated IAP and progressive renal insufficiency refractory to intensive medical therapy, mechanical fluid removal can be associated with improvements in IAP and renal function.The renal and hemodynamic profiles of 9 consecutive, volume-overloaded subjects with ADHF and elevated IAP, refractory to intensive medical therapy, were prospectively collected. All subjects experienced progressive elevation of serum creatinine and IAP in response to intravenous loop diuretics. Within 12 hours after mechanical fluid removal via paracentesis (n = 5, mean volume removed 3187 +/- 1772 mL) or ultrafiltration (n = 4, mean volume removed 1800 +/- 690 mL), there was a significant reduction in IAP (from 13 +/- 4 mm Hg to 7 +/- 2 mm Hg, P = .001), with corresponding improvement in renal function (serum creatinine from 3.4 +/- 1.4 mg/dL to 2.4 +/- 1.1 mg/dL, P = .01) without significantly altering any hemodynamic measurement.In volume-overloaded patients admitted with ADHF refractory to intensive medical therapy, we observed a reduction of otherwise persistently elevated IAP with corresponding improvement in renal function after mechanical fluid removal.
DOI: 10.1016/s0735-1097(03)00159-1
2003
Cited 153 times
Fluid retention after initiation of thiazolidinedione therapy in diabetic patients with established chronic heart failure
We sought to define the characteristics of fluid retention after thiazolidinedione (TZD) initiation in patients with established heart failure (HF).Fluid retention associated with the use of TZD is commonly attributed to exacerbation of HF, which has led to the proscription of these potentially useful agents in patients with chronic HF.We examined 111 consecutive diabetic patients with chronic systolic HF who were treated with TZD from January 1999 to June 2001. A retrospective chart review was performed to determine the incidence of fluid retention in this cohort. Physical signs of fluid retention were compared between TZD users and an age- and gender-matched control group of diabetic, non-TZD users with chronic HF who had fluid retention. Baseline clinical and echocardiographic data were compared between TZD users with and without fluid retention.Nineteen TZD users (17.1%) developed fluid retention, which reversed after drug withdrawal and presented predominantly as peripheral and not central edema. Comparing patients in the upper and lower tertiles of weight gain, more female patients and insulin users developed TZD-related fluid retention. However, there were no differences in the baseline New York Heart Association functional class or echocardiographic severity of cardiac dysfunction.Although fluid retention after treatment with TZD in diabetic patients with chronic systolic HF occurs, the mechanism is undefined. Fluid retention related to TZD tends to be peripheral and is usually reversible after drug withdrawal. No direct association between the risk of fluid retention and the baseline degree of severity of HF was observed.
DOI: 10.1161/atvbaha.112.253930
2012
Cited 147 times
Clinical and Genetic Association of Serum Paraoxonase and Arylesterase Activities With Cardiovascular Risk
Diminished serum paraoxonase and arylesterase activities (measures of paraoxonase-1 [PON-1] function) in humans have been linked to heightened systemic oxidative stress and atherosclerosis risk. The clinical prognostic use of measuring distinct PON-1 activities has not been established, and the genetic determinants of PON-1 activities are not known.We established analytically robust high-throughput assays for serum paraoxonase and arylesterase activities and measured these in 3668 stable subjects undergoing elective coronary angiography without acute coronary syndrome and were prospectively followed for major adverse cardiovascular events (MACE= death, myocardial infarction, stroke) over 3 years. Low serum arylesterase and paraoxonase activities were both associated with increased risk for MACE, with arylesterase activity showing greatest prognostic value (quartile 4 versus quartile 1; hazard ratio 2.63; 95% CI, 1.97-3.50; P<0.01). Arylesterase remained significant after adjusting for traditional risk factors, C-reactive protein, and creatinine clearance (hazard ratio, 2.20; 95% CI, 1.60-3.02; P<0.01), predicted future development of MACE in both primary and secondary prevention populations, and reclassified risk categories incrementally to traditional clinical variables. A genome-wide association study identified distinct single nucleotide polymorphisms within the PON-1 gene that were highly significantly associated with serum paraoxonase (1.18×10(-303)) or arylesterase (4.99×10(-116)) activity but these variants were not associated with either 3-year MACE risk in an angiographic cohort (n=2136) or history of either coronary artery disease or myocardial infarction in the Coronary Artery Disease Genome-Wide Replication and Meta-Analysis consortium (n≈80 000 subjects).Diminished serum arylesterase activity, but not the genetic determinants of PON-1 functional measures, provides incremental prognostic value and clinical reclassification of stable subjects at risk of developing MACE.
DOI: 10.1016/j.jacc.2007.07.094
2008
Cited 146 times
Evaluation and Long-Term Prognosis of New-Onset, Transient, and Persistent Anemia in Ambulatory Patients With Chronic Heart Failure
This study sought to determine the characteristics and long-term prognosis of anemia in ambulatory patients with chronic heart failure.Anemia is prevalent in heart failure, and may portend poor outcomes.We reviewed 6,159 consecutive outpatients with chronic stable heart failure at baseline, short-term (3-month) follow-up, and long-term (6-month) follow-up between 2001 and 2006. Clinical, demographic, laboratory, and echocardiographic data were reviewed from electronic medical records. Mortality rates were determined from 6-month follow-up to end of study period.Prevalence of anemia (hemoglobin [Hb] <12 g/dl for men, <11 g/dl for women) was 17.2% in our cohort. Diabetes, B-natriuretic peptide, left ventricular ejection fraction, and estimated glomerular filtration rate were independent predictors of baseline anemia. Documented evaluation of anemia was found in only 3% of all anemic patients, and better in internal medicine than in cardiology clinics. At 6-month follow-up, new-onset anemia developed in 16% of patients without prior anemia, whereas 43% patients with anemia at baseline had resolution of their hemoglobin levels. Higher total mortality rates were evident in patients with persistent anemia (58% vs. 31%, p < 0.0001) or with incident anemia (45% vs. 31%, p < 0.0001) compared with those with without anemia at 6 months.These observations in a broad unselected outpatient cohort suggest that anemia in patients with heart failure is under-recognized and underevaluated. However, resolution of anemia was evident in up to 43% of patients who presented initially with anemia, and did not pose greater long-term risk for all-cause mortality. However, the presence of persistent anemia conferred poorest survival in patients with heart failure when compared with that of incident, resolved, or no anemia.
DOI: 10.1016/j.jacc.2013.05.027
2013
Cited 146 times
Timing of Hemoconcentration During Treatment of Acute Decompensated Heart Failure and Subsequent Survival
This study sought to determine if the timing of hemoconcentration influences associated survival. Indicating a reduction in intravascular volume, hemoconcentration during the treatment of decompensated heart failure has been associated with reduced mortality. However, it is unclear if this survival advantage stems from the improved intravascular volume or if healthier patients are simply more responsive to diuretics. Rapid diuresis early in the hospitalization should similarly identify diuretic responsiveness, but hemoconcentration this early would not indicate euvolemia if extravascular fluid has not yet equilibrated. Consecutive admissions at a single center with a primary discharge diagnosis of heart failure were reviewed (N = 845). Hemoconcentration was defined as an increase in both hemoglobin and hematocrit levels, then further dichotomized into early or late hemoconcentration by using the midway point of the hospitalization. Hemoconcentration occurred in 422 (49.9%) patients (41.5% early and 58.5% late). Patients with late versus early hemoconcentration had similar baseline characteristics, cumulative in-hospital loop diuretic administered, and worsening of renal function. However, patients with late hemoconcentration versus early hemoconcentration had higher average daily loop diuretic doses (p = 0.001), greater weight loss (p < 0.001), later transition to oral diuretics (p = 0.03), and shorter length of stay (p < 0.001). Late hemoconcentration conferred a significant survival advantage (hazard ratio: 0.74 [95% confidence interval: 0.59 to 0.93]; p = 0.009), whereas early hemoconcentration offered no significant mortality benefit (hazard ratio: 1.0 [95% confidence interval: 0.80 to 1.3]; p = 0.93) over no hemoconcentration. Only hemoconcentration occurring late in the hospitalization was associated with improved survival. These results provide further support for the importance of achieving sustained decongestion during treatment of decompensated heart failure.
DOI: 10.1016/j.jcmg.2009.01.012
2009
Cited 133 times
Right Atrial Volume Index in Chronic Systolic Heart Failure and Prognosis
The aim of this study was to determine the relationship between right atrial volume index (RAVI) and right ventricular (RV) systolic and diastolic function, as well as long-term prognosis in patients with chronic systolic heart failure (HF). RV dysfunction is associated with poor prognosis in patients with HF, although echocardiographic assessment of RV systolic and diastolic dysfunction is challenging. The ability to visualize the RA allows a quantitative, highly reproducible assessment of the RA volume that can be indexed to body surface area. The ADEPT (Assessment of Doppler Echocardiography for Prognosis and Therapy) trial enrolled 192 subjects with chronic systolic HF (left ventricular ejection fraction [LVEF] ≤35%). The RA volume was calculated by Simpson's method using single-plane RA area and indexed to body surface area (RAVI). RV systolic function was graded as normal, mild, mild-moderate, moderate, moderately severe, or severe dysfunction. In our study cohort, the mean RAVI was 28 ± 15 ml/m2, and increased with worsening RV systolic dysfunction, LVEF, and LV diastolic dysfunction (Spearman's r = 0.61, r = 0.26, and r = 0.51, respectively; p < 0.001 for all). RAVI correlated modestly with echocardiographic estimates of RV diastolic dysfunction, including tricuspid early/late velocities ratio (Spearman's r = 0.34, p < 0.0001), hepatic vein systolic/diastolic ratio (Spearman's r = −0.26, p < 0.001) but not tricuspid early/tricuspid annular early velocities ratio (E/Ea) (Spearman's r = 0.12, p = 0.11). Increasing tertiles of RAVI were predictive of death, transplant, and/or HF hospitalization (log-rank p = 0.0002) and remained an independent predictor of adverse clinical events after adjusting for age, B-type natriuretic peptide, LV ejection fraction, RV systolic dysfunction, and tricuspid E/Ea ratio (hazard ratio: 2.00, 95% confidence interval: 1.15 to 3.58, p = 0.013). In patients with chronic systolic HF, RAVI is a determinant of right-sided systolic dysfunction. This quantitative and reproducible echocardiographic marker provides independent risk prediction of long-term adverse clinical events.
DOI: 10.1161/jaha.116.004237
2016
Cited 133 times
Trimethylamine <i>N</i> ‐Oxide and Mortality Risk in Patients With Peripheral Artery Disease
Production of the proatherogenic metabolite, trimethylamine N-oxide (TMAO), from dietary nutrients by intestinal microbiota enhances atherosclerosis development in animal models and is associated with atherosclerotic coronary artery disease in humans. The utility of studying plasma levels of TMAO to risk stratify in patients with peripheral artery disease (PAD) has not been reported.We examined the relationship between fasting plasma TMAO and all-cause mortality (5-year), stratified by subtypes of PAD and presence of coronary artery disease in 935 patients with PAD who underwent elective angiography for cardiac evaluation at a tertiary care hospital. Median plasma TMAO was 4.8 μmol/L (interquartile range, 2.9-8.0 μmol/L). Elevated TMAO levels were associated with 2.7-fold increased mortality risk (fourth versus first quartiles, hazard ratio 2.86, 95% CI 1.82-3.97, P<0.001). Following adjustments for traditional risk factors, inflammatory biomarkers, and history of coronary artery disease, the highest TMAO quartile remained predictive of 5-year mortality (adjusted hazard ratio 2.06, 95% CI 1.36-3.11, P<0.001). Similar prognostic value for elevated TMAO was seen for subjects with carotid artery, non-carotid artery, or lower extremity PAD. TMAO provided incremental prognostic value for all-cause mortality (net reclassification index, 40.22%; P<0.001) and improvement in area under receiver operator characteristic curve (65.7% versus 69.4%; P=0.013).TMAO, a pro-atherogenic metabolite formed by gut microbes, predicts long-term adverse event risk and incremental prognostic value in patients with PAD. These findings point to the potential for TMAO to help improve selection of high-risk PAD patients with or without significant coronary artery disease, who likely need more aggressive and specific dietary and pharmacologic therapy.
DOI: 10.1016/j.jacc.2013.02.005
2013
Cited 131 times
2013 ACCF/ACR/ASE/ASNC/SCCT/SCMR Appropriate Utilization of Cardiovascular Imaging in Heart Failure
Peter Alagona, MD[⁎][1] Gerard Aurigemma, MD[‡][2] Javed Butler, MD, MPH[§][3] Don Casey, MD, MPH, MBA[∥][4] Ricardo Cury, MD[#][5] Scott Flamm, MD[¶][6] Tim Gardner, MD[⁎⁎][7] Rajesh Krishnamurthy, MD[††][8] Joseph Messer, MD[⁎][1] Michael W. Rich, MD[‡‡][9] Henry
DOI: 10.1161/circheartfailure.113.000507
2014
Cited 131 times
Prevalence and Prognostic Importance of Changes in Renal Function After Mechanical Circulatory Support
The long-term durability and prognostic significance of improvement in renal function after mechanical circulatory support (MCS) has yet to be characterized in a large multicenter population. The primary goals of this analysis were to describe serial post-MCS changes in estimated glomerular filtration rate (eGFR) and determine their association with all-cause mortality.Adult patients enrolled in the Interagency Registry for Mechanically Assisted Circulatory Support (INTERMACS) with serial creatinine levels available (n=3363) were studied. Early post-MCS, eGFR improved substantially (median improvement, 48.9%; P<0.001) with 22.3% of the population improving their eGFR by ≥100% within the first few weeks. However, in the majority of patients, this improvement was transient, and by 1 year, eGFR was only 6.7% above the pre-MCS value (P<0.001). This pattern of early improvement followed by deterioration in eGFR was observed with both pulsatile and continuous-flow devices. Interestingly, poor survival was associated with both marked improvement (adjusted hazard ratio [HR], 1.64; 95% confidence interval [CI], 1.19-2.26; P=0.002) and worsening in eGFR (adjusted HR, 1.63; 95% CI, 1.15-2.13; P=0.004).Post-MCS, early improvement in renal function is common but seems to be largely transient and not necessarily indicative of an improved prognosis. This pattern was observed with both pulsatile and continuous-flow devices. Additional research is necessary to better understand the mechanistic basis for these complex post-MCS changes in renal function and their associated survival disadvantage.URL: http://www.clinicaltrials.gov. Unique identifier: NCT00119834.
DOI: 10.1093/eurheartj/ehx035
2017
Cited 130 times
Renal sodium avidity in heart failure: from pathophysiology to treatment strategies
Abstract Increased neurohumoral stimulation resulting in excessive sodium avidity and extracellular volume overload are hallmark features of decompensated heart failure. Especially in case of concomitant renal dysfunction, the kidneys often fail to elicit effective natriuresis. While assessment of renal function is generally performed by measuring serum creatinine–a surrogate for glomerular filtration-, this only represents part of the nephron’s function. Alterations in tubular sodium handling are at least equally important in the development of volume overload and congestion. Venous congestion and neurohumoral activation in advanced HF further promote renal sodium and water retention. Interestingly, early on, before clinical signs of heart failure are evident, intrinsic renal derangements already impair natriuresis. This clinical review discusses the importance of heart failure (HF) induced changes in different nephron segments. A better understanding of cardiorenal interactions which ultimately result in sodium avidity in HF might help to treat and prevent congestion in chronic and acute HF.
DOI: 10.1016/j.jacc.2012.07.047
2012
Cited 129 times
Incremental Prognostic Value of Assessing Left Ventricular Myocardial Mechanics in Patients With Chronic Systolic Heart Failure
This study sought to examine the ability of left ventricular (LV) global longitudinal strain (GLS) to assess disease severity in patients with chronic systolic heart failure (HF).Left ventricular GLS is a sensitive measure of LV mechanics. Its relationship with standard clinical markers and long-term adverse events in chronic systolic HF is not well established.In 194 chronic systolic HF patients, we performed comprehensive echocardiography with assessment of GLS by velocity vector imaging averaged from apical 4-chamber and 2-chamber views. Death, cardiac transplantation, and HF hospitalization were tracked for 5 years.In our study cohort (age 57 ± 14 years, left ventricular ejection fraction [LVEF] 26 ± 6%, median N-terminal pro-B-type natriuretic peptide [NT-proBNP] 1,158 pg/ml), the mean GLS was -7.1 ± 3.3%. The GLS worsened with increasing New York Heart Association functional class (rank-sum p < 0.0001) and higher NT-proBNP (r = 0.42, p < 0.0001). The GLS correlated with LV cardiac structure (LV mass index: r = 0.35, p < 0.0001; LV end-diastolic volume index: r = 0.43, p < 0.0001) and LVEF (r = -0.66, p < 0.0001). A lower magnitude of GLS was associated with worsening LV diastolic function (E/e' septal: r = 0.33, p < 0.0001), right ventricular (RV) systolic function (RV s': r = -0.30, p < 0.0001), and RV diastolic function (RV e'/a': r = 0.16, p = 0.033). GLS predicted long-term adverse events (hazard ratio: 1.55, 95% confidence interval: 1.21 to 2.00; p < 0.001). Worsening strain (GLS ≥-6.95%) predicted adverse events after adjustment for age, sex, ischemic etiology, E/e' septal, and NT-proBNP (hazard ratio: 2.04, 95% confidence interval: 1.09 to 3.94; p = 0.025) and age, sex, ischemic etiology, and LVEF (hazard ratio: 2.15, 95% confidence interval: 1.19 to 4.02; p = 0.011).In chronic systolic HF, worsening LV GLS is associated with more severe LV diastolic dysfunction and RV systolic and diastolic dysfunction, and provides incremental prognostic value to LVEF.
DOI: 10.1002/ejhf.35
2013
Cited 126 times
The kidney in congestive heart failure: ‘are natriuresis, sodium, and diuretics really the good, the bad and the ugly?’
This review discusses renal sodium handling in heart failure. Increased sodium avidity and tendency to extracellular volume overload, i.e. congestion, are hallmark features of the heart failure syndrome. Particularly in the case of concomitant renal dysfunction, the kidneys often fail to elicit potent natriuresis. Yet, assessment of renal function is generally performed by measuring serum creatinine, which has inherent limitations as a biomarker for the glomerular filtration rate (GFR). Moreover, glomerular filtration only represents part of the nephron's function. Alterations in the fractional reabsorptive rate of sodium are at least equally important in emerging therapy-refractory congestion. Indeed, renal blood flow decreases before the GFR is affected in congestive heart failure. The resulting increased filtration fraction changes Starling forces in peritubular capillaries, which drive sodium reabsorption in the proximal tubules. Congestion further stimulates this process by augmenting renal lymph flow. Consequently, fractional sodium reabsorption in the proximal tubules is significantly increased, limiting sodium delivery to the distal nephron. Orthosympathetic activation probably plays a pivotal role in those deranged intrarenal haemodynamics, which ultimately enhance diuretic resistance, stimulate neurohumoral activation with aldosterone breakthrough, and compromise the counter-regulatory function of natriuretic peptides. Recent evidence even suggests that intrinsic renal derangements might impair natriuresis early on, before clinical congestion or neurohumoral activation are evident. This represents a paradigm shift in heart failure pathophysiology, as it suggests that renal dysfunction-although not by conventional GFR measurements-is driving disease progression. In this respect, a better understanding of renal sodium handling in congestive heart failure is crucial to achieve more tailored decongestive therapy, while preserving renal function.
DOI: 10.1016/j.cardfail.2014.03.006
2014
Cited 125 times
Insufficient Natriuretic Response to Continuous Intravenous Furosemide Is Associated With Poor Long-Term Outcomes in Acute Decompensated Heart Failure
Treatment of acute decompensated heart failure (ADHF) with loop diuretics, such as furosemide, is frequently complicated by insufficient urine sodium excretion. We hypothesize that insufficient natriuretic response to diuretic therapy, characterized by lower urine sodium (UNa) and urine furosemide, is associated with subsequent inadequate decongestion, worsening renal function, and adverse long term events.We enrolled 52 consecutive patients with ADHF and measured serum and urine sodium (UNa), urine creatinine (UCr), and urine furosemide (UFurosemide) levels on a spot sample taken after treatment with continuous intravenous furosemide, and followed clinical and renal variables as well as adverse long-term clinical outcomes (death, rehospitalizations, and cardiac transplantation). We observed similar correlations between UNa:UFurosemide ratio and UNa and fractional excretion of sodium (FENa) with 24-hour net urine output (r = 0.52-0.64, all P < .01) and 24-hour weight loss (r = 0.44-0.56; all P < .01). Interestingly, FENa (but not UNa or UNa:UFurosemide) were influenced by estimated glomerular filtration rate (eGFR). We observed an association between lower UNa:UFurosemide with greater likelihood of worsening renal function (hazard ratio [HR] 3.01; P = .02) and poorer adverse clinical outcomes (HR 1.63, P = .008) after adjusting for age and eGFR. Meanwhile, both diminished weight loss and net fluid output over 24 hours of continuous intravenous furosemide were observed when UNa:UFurosemide ratios were <2 mmol/mg or when UNa <50 mmol.In patients with ADHF receiving continuous furosemide infusion, impaired natriuretic response to furosemide is associated with greater likelihood of worsening renal function and future adverse long-term outcomes, independently from and incrementally with decreasing intrinsic glomerular filtration.
DOI: 10.1161/circheartfailure.112.968511
2012
Cited 125 times
Prognostic Role of Pulmonary Arterial Capacitance in Advanced Heart Failure
Right ventricular (RV) dysfunction frequently occurs and independently prognosticates in left-sided heart failure. It is not clear which RV afterload measure has the greatest impact on RV function and prognosis. We examined the determinants, prognostic role, and response to treatment of pulmonary arterial capacitance (PAC, ratio of stroke volume over pulmonary pulse pressure), in relation to pulmonary vascular resistance (PVR) in heart failure.We reviewed 724 consecutive patients with heart failure who underwent right heart catheterization between 2000 and 2005. Changes in PAC were explored in an independent cohort of 75 subjects treated for acute decompensated heart failure. PAC showed a strong inverse relation with PVR (r=-0.64) and wedge pressure (r=-0.73), and provides stronger prediction of significant RV failure than PVR (area under the curve ROC 0.74 versus 0.67, respectively, P=0.003). During a mean follow-up of 3.2±2.2 years, both lower PAC (P<0.0001) and higher PVR (P<0.0001) portend more adverse clinical events (all-cause mortality and cardiac transplantation). In multivariate analysis, PAC (but not PVR) remains an independent predictor (Hazard ratio=0.92 [95% CI: 0.84-1.0, P=0.037]). Treatment of heart failure resulted in a decrease in PVR (270±165 to 211±88 dynes·s(-1)·cm(-5), P=0.002), a larger increase in PAC (1.65±0.64 to 2.61±1.42 mL/mm Hg, P<0.0001), leading to an increase in pulmonary arterial time constant (PVR×PAC) (0.29±0.12 to 0.37±0.15 second, P<0.0001).PAC bundles the effects of PVR and left-sided filling pressures on RV afterload, explaining its strong relation with RV dysfunction, poor long-term prognosis, and response to therapy.
DOI: 10.1016/j.jacc.2015.06.007
2015
Cited 123 times
Prognostic Role of Serum Chloride Levels in Acute Decompensated Heart Failure
Acute decompensated heart failure (ADHF) can be complicated by electrolyte abnormalities, but the major focus has been concentrated on the clinical significance of serum sodium levels.This study sought to determine the prognostic significance of serum chloride levels in relation to serum sodium levels in patients with ADHF.We reviewed 1,318 consecutive patients with chronic heart failure admitted for ADHF to the Cleveland Clinic between July 2008 and December 2013. We also validated our findings in an independent ADHF cohort from the University of Pennsylvania (n = 876).Admission serum chloride levels during hospitalization for ADHF were independently and inversely associated with long-term mortality (hazard ratio [HR] per unit change: 0.94; 95% confidence interval [CI]: 0.92 to 0.95; p < 0.001). After multivariable risk adjustment, admission chloride levels remained independently associated with mortality (HR per unit change: 0.93; 95% CI: 0.90 to 0.97; p < 0.001) in contrast to admission sodium levels, which were no longer significant (p > 0.05). Results were similar in the validation cohort in unadjusted (HR per unit change for mortality risk within 1 year: 0.93; 95% CI: 0.91 to 0.95; p < 0.001) and multivariable risk-adjusted analysis (HR per unit change for mortality risk within 1 year: 0.95; 95% CI: 0.92 to 0.99; p = 0.01).These observations in a contemporary advanced ADHF cohort suggest that serum chloride levels at admission are independently and inversely associated with mortality. The prognostic value of serum sodium in ADHF was diminished compared with chloride.
DOI: 10.1681/asn.2012030254
2013
Cited 123 times
Protein Carbamylation Predicts Mortality in ESRD
Traditional risk factors fail to explain the increased risk for cardiovascular morbidity and mortality in ESRD. Cyanate, a reactive electrophilic species in equilibrium with urea, posttranslationally modifies proteins through a process called carbamylation, which promotes atherosclerosis. The plasma level of protein-bound homocitrulline (PBHCit), which results from carbamylation, predicts major adverse cardiac events in patients with normal renal function, but whether this relationship is similar in ESRD is unknown. We quantified serum PBHCit in a cohort of 347 patients undergoing maintenance hemodialysis with 5 years of follow-up. Kaplan-Meier analyses revealed a significant association between elevated PBHCit and death (log-rank P<0.01). After adjustment for patient characteristics, laboratory values, and comorbid conditions, the risk for death among patients with PBHCit values in the highest tertile was more than double the risk among patients with values in the middle tertile (adjusted hazard ratio [HR], 2.4; 95% confidence interval [CI], 1.5-3.9) or the lowest tertile (adjusted HR, 2.3; 95% CI, 1.5-3.7). Including PBHCit significantly improved the multivariable model, with a net reclassification index of 14% (P<0.01). In summary, serum PBHCit, a footprint of protein carbamylation, predicts increased cardiovascular risk in patients with ESRD, supporting a mechanistic link among uremia, inflammation, and atherosclerosis.