1
|
Benedetti S, Gemma Nasoni M, Palma F, Citarella R, Luchetti F. Serum changes in sTWEAK and its scavenger receptor sCD163 in ultramarathon athletes running the 24-h race. Cytokine 2020; 137:155315. [PMID: 33011401 DOI: 10.1016/j.cyto.2020.155315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/28/2020] [Accepted: 09/22/2020] [Indexed: 11/17/2022]
Abstract
In the present investigation, the serum changes of sTWEAK levels, a multifunctional cytokine involved in tissue response to acute injury and inflammation, and of its scavenger receptor sCD163, were monitored for the first time in ultramarathon athletes running the 24-h competition, an extremely demanding race in terms of muscular and physiological exertion. To this aim, venous blood samples were collected from each participant (n = 22, M = 12, F = 10) both before and immediately after the 24-h running. Other than sTWEAK and sCD163, the common serum biomarkers of inflammation (namely CRP and IL-6) and tissue injury (such as CPK, LDH, CPK-MB, troponin-I, and NT-proBNP) were evaluated. All parameters were within the reference ranges at baseline, indicating no alterations of the normal physiological processes before the competition; on the contrary, most biomarkers of tissue damage and inflammation strongly increased after the ultramarathon race. Interestingly, a significant decrement of sTWEAK levels associated with an increment of its scavenger receptor sCD163 was observed at post-race. Positive relationships were evidenced between IL-6 and sCD163 levels and the markers of cardiac damage troponin-I and NT-proBNP. On the contrary, sTWEAK showed an inverse correlation with IL-6 and NT-proBNP. This study opens the way to further investigations aimed at clarifying the role of TWEAK pathway during the prolonged ultraendurance activity, paying particular attention to the link of IL-6, CD163 and TWEAK with the cardiac function.
Collapse
Affiliation(s)
- Serena Benedetti
- Department of Biomolecular Sciences, Section of Biochemistry and Biotechnology, University of Urbino Carlo Bo, Urbino, Italy.
| | - Maria Gemma Nasoni
- Department of Biomolecular Sciences, Section of Morphology, Physiology and Environmental Biology, University of Urbino Carlo Bo, Urbino, Italy
| | - Francesco Palma
- Department of Biomolecular Sciences, Section of Biochemistry and Biotechnology, University of Urbino Carlo Bo, Urbino, Italy
| | | | - Francesca Luchetti
- Department of Biomolecular Sciences, Section of Morphology, Physiology and Environmental Biology, University of Urbino Carlo Bo, Urbino, Italy
| |
Collapse
|
2
|
Espinoza-Derout J, Hasan KM, Shao XM, Jordan MC, Sims C, Lee DL, Sinha S, Simmons Z, Mtume N, Liu Y, Roos KP, Sinha-Hikim AP, Friedman TC. Chronic intermittent electronic cigarette exposure induces cardiac dysfunction and atherosclerosis in apolipoprotein-E knockout mice. Am J Physiol Heart Circ Physiol 2019; 317:H445-H459. [PMID: 31172811 DOI: 10.1152/ajpheart.00738.2018] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Electronic cigarettes (e-cigarettes), also known as electronic nicotine delivery systems, are a popular alternative to conventional nicotine cigarettes, both among smokers and those who have never smoked. In spite of the widespread use of e-cigarettes and the proposed detrimental cardiac and atherosclerotic effects of nicotine, the effects of e-cigarettes on these systems are not known. In this study, we investigated the cardiovascular and cardiac effects of e-cigarettes with and without nicotine in apolipoprotein-E knockout (ApoE-/-) mice. We developed an e-cigarette exposure model that delivers nicotine in a manner similar to that of human e-cigarettes users. Using commercially available e-cigarettes, bluCig PLUS, ApoE-/- mice were exposed to saline, e-cigarette without nicotine [e-cigarette (0%)], and e-cigarette with 2.4% nicotine [e-cigarette (2.4%)] aerosol for 12 wk. Echocardiographic data show that mice treated with e-cigarette (2.4%) had decreased left ventricular fractional shortening and ejection fraction compared with e-cigarette (0%) and saline. Ventricular transcriptomic analysis revealed changes in genes associated with metabolism, circadian rhythm, and inflammation in e-cigarette (2.4%)-treated ApoE-/- mice. Transmission electron microscopy revealed that cardiomyocytes of mice treated with e-cigarette (2.4%) exhibited ultrastructural abnormalities indicative of cardiomyopathy. Additionally, we observed increased oxidative stress and mitochondrial DNA mutations in mice treated with e-cigarette (2.4%). ApoE-/- mice on e-cigarette (2.4%) had also increased atherosclerotic lesions compared with saline aerosol-treated mice. These results demonstrate adverse effects of e-cigarettes on cardiac function in mice.NEW & NOTEWORTHY The present study is the first to show that mice exposed to nicotine electronic cigarettes (e-cigarettes) have decreased cardiac fractional shortening and ejection fraction in comparison with controls. RNA-seq analysis reveals a proinflammatory phenotype induced by e-cigarettes with nicotine. We also found increased atherosclerosis in the aortic root of mice treated with e-cigarettes with nicotine. Our results show that e-cigarettes with nicotine lead to detrimental effects on the heart that should serve as a warning to e-cigarette users and agencies that regulate them.
Collapse
Affiliation(s)
- Jorge Espinoza-Derout
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Kamrul M Hasan
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Xuesi M Shao
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California.,David Geffen School of Medicine at University of California, Los Angeles, California
| | - Maria C Jordan
- David Geffen School of Medicine at University of California, Los Angeles, California
| | - Carl Sims
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Desean L Lee
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Satyesh Sinha
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California.,David Geffen School of Medicine at University of California, Los Angeles, California
| | - Zena Simmons
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Norma Mtume
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California
| | - Yanjun Liu
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California.,David Geffen School of Medicine at University of California, Los Angeles, California
| | - Kenneth P Roos
- David Geffen School of Medicine at University of California, Los Angeles, California
| | - Amiya P Sinha-Hikim
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California.,David Geffen School of Medicine at University of California, Los Angeles, California
| | - Theodore C Friedman
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, California.,David Geffen School of Medicine at University of California, Los Angeles, California
| |
Collapse
|
3
|
Tang B, Zhong Z, Qiu Z, Wu HP, Hu JY, Ma JP, Wu JP. Serum soluble TWEAK levels in severe traumatic brain injury and its prognostic significance. Clin Chim Acta 2019; 495:227-232. [PMID: 31009601 DOI: 10.1016/j.cca.2019.04.070] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/13/2019] [Accepted: 04/17/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND Severe traumatic brain injury (sTBI) is characterized by a high mortality. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) participates in inflammation. We determined serum soluble TWEAK (sTWEAK) levels with respect to its prognostic ability. METHODS This was a single-center prospective, observational study that was performed from December 2014 to December 2017. A total of 114 sTBI patients who met the inclusion criteria and 114 randomly selected healthy controls were included in the study. Serum sTWEAK levels were gauged. Patients were followed-up until death or completion of 6 months. Poor outcome was referred to as Glasgow outcome scale score of 1-3. RESULTS In comparison with controls, patients displayed predominantly higher serum sTWEAK levels. Serum sTWEAK levels were strongly correlated with Glasgow coma scale scores and serum C-reactive protein levels. 32 patients (28.1%) died and 60 patients (52.6%) suffered from a poor outcome. Receiver operating characteristic curve analysis clearly showed that serum sTWEAK levels had substantially high predictive performance for 6-month mortality and poor outcome. Serum sTWEAK emerged as an independent predictor for 6-month mortality, overall survival and poor outcome. CONCLUSIONS Raised serum sTWEAK levels are closely related to increasing inflammatory response, elevated trauma severity and worse clinical outcome after sTBI.
Collapse
Affiliation(s)
- Bei Tang
- Department of Critical Care Medicine, The First People's Hospital of Jiande City, 599 Yanzhou Main Road, Jiande 311600, China
| | - Ze Zhong
- Department of Critical Care Medicine, The First People's Hospital of Jiande City, 599 Yanzhou Main Road, Jiande 311600, China.
| | - Zheng Qiu
- Department of Neurosurgery, The First People's Hospital of Jiande City, 599 Yanzhou Main Road, Jiande 311600, China
| | - Hui-Ping Wu
- Department of Critical Care Medicine, The First People's Hospital of Jiande City, 599 Yanzhou Main Road, Jiande 311600, China
| | - Jia-Yuan Hu
- Department of Critical Care Medicine, The First People's Hospital of Jiande City, 599 Yanzhou Main Road, Jiande 311600, China
| | - Jian-Ping Ma
- Department of Critical Care Medicine, The First People's Hospital of Jiande City, 599 Yanzhou Main Road, Jiande 311600, China
| | - Jin-Ping Wu
- Department of Critical Care Medicine, The First People's Hospital of Jiande City, 599 Yanzhou Main Road, Jiande 311600, China
| |
Collapse
|
4
|
Das NA, Carpenter AJ, Yoshida T, Kumar SA, Gautam S, Mostany R, Izadpanah R, Kumar A, Mummidi S, Siebenlist U, Chandrasekar B. TRAF3IP2 mediates TWEAK/TWEAKR-induced pro-fibrotic responses in cultured cardiac fibroblasts and the heart. J Mol Cell Cardiol 2018; 121:107-123. [PMID: 29981796 DOI: 10.1016/j.yjmcc.2018.07.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/20/2018] [Accepted: 07/03/2018] [Indexed: 12/19/2022]
Abstract
Persistent inflammation promotes development and progression of heart failure (HF). TWEAK (TNF-Related WEAK Inducer Of Apoptosis), a NF-κB- and/or AP-1-responsive proinflammatory cytokine that signals via TWEAK receptor (TWEAKR), is expressed at high levels in human and preclinical models of HF. Since the adapter molecule TRAF3IP2 (TRAF3 Interacting Protein 2) is an upstream regulator of various proinflammatory pathways, including those activated by NF-κB and AP-1, we hypothesized that targeting TRAF3IP2 inhibits TWEAK-induced proinflammatory and pro-fibrotic responses in vitro and in vivo. Consistent with the hypothesis, forced expression of TRAF3IP2 upregulated TWEAK and its receptor expression in cultured adult mouse cardiac fibroblasts (CF). Further, exogenous TWEAK upregulated TRAF3IP2 expression in a time- and dose-dependent manner, suggesting a positive-feedback regulation of TRAF3IP2 and TWEAK. TWEAK also promoted TRAF3IP2 nuclear translocation. Confirming its critical role in TWEAK signaling, silencing TRAF3IP2 inhibited TWEAK autoregulation, TWEAKR upregulation, p38 MAPK, NF-κB and AP-1 activation, inflammatory cytokine expression, MMP and TIMP1 activation, collagen expression and secretion, and importantly, proliferation and migration. Recapitulating these in vitro results, continuous infusion of TWEAK for 7 days increased systolic blood pressure (SBP), upregulated TRAF3IP2 expression, activated p38 MAPK, NF-κB and AP-1, induced the expression of multiple proinflammatory and pro-fibrotic mediators, and interstitial fibrosis in hearts of wild type mice. These proinflammatory and pro-fibrotic changes occurred in conjunction with myocardial hypertrophy and contractile dysfunction. Importantly, genetic ablation of TRAF3IP2 inhibited these TWEAK-induced adverse cardiac changes independent of increases in SBP, indicating that TRAF3IP2 plays a causal role, and thus a therapeutic target, in chronic inflammatory and fibro-proliferative diseases.
Collapse
Affiliation(s)
- Nitin A Das
- Cardiothoracic Surgery, University of Texas Health Science Center, San Antonio, TX, USA
| | - Andrea J Carpenter
- Cardiothoracic Surgery, University of Texas Health Science Center, San Antonio, TX, USA
| | - Tadashi Yoshida
- Medicine/Cardiology, University of Missouri School of Medicine, Columbia, MO, USA
| | - Senthil A Kumar
- Medicine/Cardiology, University of Missouri School of Medicine, Columbia, MO, USA
| | - Sandeep Gautam
- Medicine/Cardiology, University of Missouri School of Medicine, Columbia, MO, USA
| | - Ricardo Mostany
- Department of Pharmacology, Tulane University Health Science Center, New Orleans, LA, USA
| | - Reza Izadpanah
- Medicine/Heart and Vascular Institute, Tulane University Health Science Center, New Orleans, LA, USA
| | - Ashok Kumar
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Srinivas Mummidi
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, The University of Texas Rio Grande Valley School of Medicine, Edinburg, TX, USA
| | | | - Bysani Chandrasekar
- Medicine/Cardiology, University of Missouri School of Medicine, Columbia, MO, USA; Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
5
|
Chen X, Farrokhi V, Singh P, Ocana MF, Patel J, Lin LL, Neubert H, Brodfuehrer J. Biomeasures and mechanistic modeling highlight PK/PD risks for a monoclonal antibody targeting Fn14 in kidney disease. MAbs 2017; 10:62-70. [PMID: 29190188 DOI: 10.1080/19420862.2017.1398873] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Discovery of the upregulation of fibroblast growth factor-inducible-14 (Fn14) receptor following tissue injury has prompted investigation into biotherapeutic targeting of the Fn14 receptor for the treatment of conditions such as chronic kidney diseases. In the development of monoclonal antibody (mAb) therapeutics, there is an increasing trend to use biomeasures combined with mechanistic pharmacokinetic/pharmacodynamic (PK/PD) modeling to enable decision making in early discovery. With the aim of guiding preclinical efforts on designing an antibody with optimized properties, we developed a mechanistic site-of-action (SoA) PK/PD model for human application. This model incorporates experimental biomeasures, including concentration of soluble Fn14 (sFn14) in human plasma and membrane Fn14 (mFn14) in human kidney tissue, and turnover rate of human sFn14. Pulse-chase studies using stable isotope-labeled amino acids and mass spectrometry indicated the sFn14 half-life to be approximately 5 hours in healthy volunteers. The biomeasures (concentration, turnover) of sFn14 in plasma reveals a significant hurdle in designing an antibody against Fn14 with desired characteristics. The projected dose (>1 mg/kg/wk for 90% target coverage) derived from the human PK/PD model revealed potential high and frequent dosing requirements under certain conditions. The PK/PD model suggested a unique bell-shaped relationship between target coverage and antibody affinity for anti-Fn14 mAb, which could be applied to direct the antibody engineering towards an optimized affinity. This investigation highlighted potential applications, including assessment of PK/PD risks during early target validation, human dose prediction and drug candidate optimization.
Collapse
Affiliation(s)
- Xiaoying Chen
- a Department of Biomedicine Design , Pfizer Inc , Cambridge , MA , United States of America
| | - Vahid Farrokhi
- b Department of Biomedicine Design , Pfizer Inc , Andover , MA , United States of America
| | - Pratap Singh
- b Department of Biomedicine Design , Pfizer Inc , Andover , MA , United States of America
| | - Mireia Fernandez Ocana
- b Department of Biomedicine Design , Pfizer Inc , Andover , MA , United States of America
| | - Jenil Patel
- b Department of Biomedicine Design , Pfizer Inc , Andover , MA , United States of America
| | - Lih-Ling Lin
- c Inflammation and Immunology Research Unit , Pfizer Inc. , Cambridge , MA , United States of America
| | - Hendrik Neubert
- b Department of Biomedicine Design , Pfizer Inc , Andover , MA , United States of America
| | - Joanne Brodfuehrer
- a Department of Biomedicine Design , Pfizer Inc , Cambridge , MA , United States of America
| |
Collapse
|
6
|
Tuttolomondo A, Simonetta I, Pinto A. MicroRNA and receptor mediated signaling pathways as potential therapeutic targets in heart failure. Expert Opin Ther Targets 2016; 20:1287-1300. [PMID: 27409295 DOI: 10.1080/14728222.2016.1212017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Cardiac remodelling is a complex pathogenetic pathway involving genome expression, molecular, cellular, and interstitial changes that cause changes in size, shape and function of the heart after cardiac injury. Areas covered: We will review recent advances in understanding the role of several receptor-mediated signaling pathways and micro-RNAs, in addition to their potential as candidate target pathways in the pathogenesis of heart failure. The myocyte is the main target cell involved in the remodelling process via ischemia, cell necrosis and apoptosis (by means of various receptor pathways), and other mechanisms mediated by micro-RNAs. We will analyze the role of some receptor mediated signaling pathways such as natriuretic peptides, mediators of glycogen synthase kinase 3 and ERK1/2 pathways, beta-adrenergic receptor subtypes and relaxin receptor signaling mechanisms, TNF/TNF receptor family and TWEAK/Fn14 axis, and some micro-RNAs as candidate target pathways in pathogenesis of heart failure. These mediators of receptor-mediated pathways and micro-RNA are the most addressed targets of emerging therapies in modern heart failure treatment strategies. Expert opinion: Future treatment strategies should address mediators involved in multiple steps within heart failure pathogenetic pathways.
Collapse
Affiliation(s)
- Antonino Tuttolomondo
- a U.O.C di Medicina Interna con Stroke Care, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S) , University of Palermo , Palermo , Italy
| | - Irene Simonetta
- a U.O.C di Medicina Interna con Stroke Care, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S) , University of Palermo , Palermo , Italy
| | - Antonio Pinto
- a U.O.C di Medicina Interna con Stroke Care, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S) , University of Palermo , Palermo , Italy
| |
Collapse
|
7
|
Soluble Fn14 Is Detected and Elevated in Mouse and Human Kidney Disease. PLoS One 2016; 11:e0155368. [PMID: 27171494 PMCID: PMC4865213 DOI: 10.1371/journal.pone.0155368] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/27/2016] [Indexed: 01/01/2023] Open
Abstract
The cytokine TWEAK and its cognate receptor Fn14 are members of the TNF/TNFR superfamily and are upregulated in tissue injury to mediate local tissue responses including inflammation and tissue remodeling. We found that in various models of kidney disease, Fn14 expression (mRNA and protein) is upregulated in the kidney. These models include: lupus nephritis mouse models (Nephrotoxic serum Transfer Nephritis and MRL.Faslpr/lpr), acute kidney injury models (Ischemia reperfusion injury and Folic acid injury), and a ZSF-1 diabetic nephropathy rat model. Fn14 expression levels correlate with disease severity as measured by disease histology. We have also shown for the first time the detection of soluble Fn14 (sFn14) in the urine and serum of mice. Importantly, we found the sFn14 levels are markedly increased in the diseased mice and are correlated with disease biomarkers including proteinuria and MCP-1. We have also detected sFn14 in human plasma and urine. Moreover, sFn14 levels, in urine are significantly increased in DN patients and correlated with proteinuria and MCP-1 levels. Thus our data not only confirm the up-regulation of Fn14/TWEAK pathway in kidney diseases, but also suggest a novel mechanism for its regulation by the generation of sFn14. The correlation of sFn14 levels and disease severity suggest that sFn14 may serve as a potential biomarker for both acute and chronic kidney diseases.
Collapse
|
8
|
Zheng Y, Chen H, Li X, Sun Y. Pay attention to cardiac remodeling in cancer cachexia. Support Care Cancer 2016; 24:3253-9. [PMID: 27108265 DOI: 10.1007/s00520-016-3222-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 04/17/2016] [Indexed: 01/21/2023]
Abstract
Cancer cachexia is a complex and multifaceted disease state characterized by fatigue, weakness, and loss of skeletal muscle and adipose tissue. Recently, the profound negative effects of cancer cachexia on cardiac tissue draw much attention, which is likely to contribute to mortality in tumor-bearing animals. The mechanism of cardiac remodeling is not so clear and involved with a series of molecular alterations. In cancer cachexia model, progressive loss of left ventricular mass and decrease in myocardial function is observed and cardiac autonomic functions are altered. Levels of several emerging cardiovascular neurohormones are found elevating in patients with cancer, but it is still controversial whether the changes could reflect the heart injury accurately. The remedy for cardiac remodeling has been explored. It is showed that exercise can modulate signaling pathways activated by wasting cytokines and impact on the resulting outcomes on heart adaptation. Some drugs, such as bisoprolol, spironolactone, perindopril, tandospirone, and simvastatin, can mitigate adverse effects of the tumor on the heart and prolong survival.
Collapse
Affiliation(s)
- Yawen Zheng
- Department of Oncology, Jinan Central Hospital, Shandong University, No. 105, Jie Fang Road, Jinan, Shandong, 250013, People's Republic of China
| | - Han Chen
- Soochow University College of Medicine, Suzhou, Jiangsu, 215000, China
| | - Xiaoqing Li
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310029, China
| | - Yuping Sun
- Department of Oncology, Jinan Central Hospital, Shandong University, No. 105, Jie Fang Road, Jinan, Shandong, 250013, People's Republic of China.
| |
Collapse
|
9
|
Nowakowski A, Walczak P, Janowski M, Lukomska B. Genetic Engineering of Mesenchymal Stem Cells for Regenerative Medicine. Stem Cells Dev 2015; 24:2219-42. [PMID: 26140302 DOI: 10.1089/scd.2015.0062] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs), which can be obtained from various organs and easily propagated in vitro, are one of the most extensively used types of stem cells and have been shown to be efficacious in a broad set of diseases. The unique and highly desirable properties of MSCs include high migratory capacities toward injured areas, immunomodulatory features, and the natural ability to differentiate into connective tissue phenotypes. These phenotypes include bone and cartilage, and these properties predispose MSCs to be therapeutically useful. In addition, MSCs elicit their therapeutic effects by paracrine actions, in which the metabolism of target tissues is modulated. Genetic engineering methods can greatly amplify these properties and broaden the therapeutic capabilities of MSCs, including transdifferentiation toward diverse cell lineages. However, cell engineering can also affect safety and increase the cost of therapy based on MSCs; thus, the advantages and disadvantages of these procedures should be discussed. In this review, the latest applications of genetic engineering methods for MSCs with regenerative medicine purposes are presented.
Collapse
Affiliation(s)
- Adam Nowakowski
- 1 NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences , Warsaw, Poland
| | - Piotr Walczak
- 2 Division of Magnetic Resonance Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine , Baltimore, Maryland.,3 Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland.,4 Department of Radiology, Faculty of Medical Sciences, University of Warmia and Mazury , Olsztyn, Poland
| | - Miroslaw Janowski
- 1 NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences , Warsaw, Poland .,2 Division of Magnetic Resonance Research, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine , Baltimore, Maryland.,3 Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Barbara Lukomska
- 1 NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences , Warsaw, Poland
| |
Collapse
|
10
|
Ren M, Li X, Hao L, Zhong J. Role of tumor necrosis factor alpha in the pathogenesis of atrial fibrillation: A novel potential therapeutic target? Ann Med 2015; 47:316-24. [PMID: 25982799 DOI: 10.3109/07853890.2015.1042030] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia in clinical practice and a major cause of morbidity and mortality. Although the fundamental mechanisms underlying AF remain incompletely understood, atrial remodeling, including structural, electrical, contractile, and autonomic remodeling, has been demonstrated to contribute to the substrate for AF maintenance. Accumulating evidence shows that tumor necrosis factor alpha (TNF-α) plays exceedingly important roles in atrial remodeling. This article reviews recent advances in the roles of TNF-α in the pathogenesis of AF, elucidates the related mechanisms, and exploits its potential usefulness as a novel therapeutic target.
Collapse
Affiliation(s)
- Manyi Ren
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital of Shandong University , China
| | | | | | | |
Collapse
|
11
|
Ghavami S, Cunnington RH, Gupta S, Yeganeh B, Filomeno KL, Freed DH, Chen S, Klonisch T, Halayko AJ, Ambrose E, Singal R, Dixon IMC. Autophagy is a regulator of TGF-β1-induced fibrogenesis in primary human atrial myofibroblasts. Cell Death Dis 2015; 6:e1696. [PMID: 25789971 PMCID: PMC4385916 DOI: 10.1038/cddis.2015.36] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 01/09/2015] [Accepted: 01/16/2015] [Indexed: 01/07/2023]
Abstract
Transforming growth factor-β1 (TGF-β1) is an important regulator of fibrogenesis in heart disease. In many other cellular systems, TGF-β1 may also induce autophagy, but a link between its fibrogenic and autophagic effects is unknown. Thus we tested whether or not TGF-β1-induced autophagy has a regulatory function on fibrosis in human atrial myofibroblasts (hATMyofbs). Primary hATMyofbs were treated with TGF-β1 to assess for fibrogenic and autophagic responses. Using immunoblotting, immunofluorescence and transmission electron microscopic analyses, we found that TGF-β1 promoted collagen type Iα2 and fibronectin synthesis in hATMyofbs and that this was paralleled by an increase in autophagic activation in these cells. Pharmacological inhibition of autophagy by bafilomycin-A1 and 3-methyladenine decreased the fibrotic response in hATMyofb cells. ATG7 knockdown in hATMyofbs and ATG5 knockout (mouse embryonic fibroblast) fibroblasts decreased the fibrotic effect of TGF-β1 in experimental versus control cells. Furthermore, using a coronary artery ligation model of myocardial infarction in rats, we observed increases in the levels of protein markers of fibrosis, autophagy and Smad2 phosphorylation in whole scar tissue lysates. Immunohistochemistry for LC3β indicated the localization of punctate LC3β with vimentin (a mesenchymal-derived cell marker), ED-A fibronectin and phosphorylated Smad2. These results support the hypothesis that TGF-β1-induced autophagy is required for the fibrogenic response in hATMyofbs.
Collapse
Affiliation(s)
- S Ghavami
- 1] Department of Physiology, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada [2] Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada [3] Department of Physiology and Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada [4] Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - R H Cunnington
- Department of Physiology and Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - S Gupta
- Department of Physiology and Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - B Yeganeh
- 1] Department of Physiology, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada [2] Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada [3] Department of Physiology and Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - K L Filomeno
- Department of Physiology and Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - D H Freed
- Department of Physiology and Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - S Chen
- Department of Physiology and Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - T Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - A J Halayko
- 1] Department of Physiology, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada [2] Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada [3] Department of Internal Medicine, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
| | - E Ambrose
- Department of Physiology and Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - R Singal
- Cardiac Sciences Program, St. Boniface General Hospital, Winnipeg, Manitoba, Canada
| | - I M C Dixon
- Department of Physiology and Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
12
|
Hernández-Romero D, Jover E, Martínez CM, Andreu-Cayuelas JM, Orenes-Piñero E, Romero-Aniorte AI, Casas T, Cánovas S, Montero-Argudo JA, Valdés M, de la Morena G, Marín F. TWEAK and NT-proBNP levels predict exercise capacity in hypertrophic cardiomyopathy. Eur J Clin Invest 2015; 45:179-86. [PMID: 25524713 DOI: 10.1111/eci.12394] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 12/14/2014] [Indexed: 01/10/2023]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is characterized by inappropriate hypertrophy, myocyte disarray and increased interstitial fibrosis. The tumour necrosis factor-like weak inducer of apoptosis (TWEAK) is a cell surface cytokine with biological activities including stimulation of cell growth, induction of inflammatory cytokines and stimulation of apoptosis. There are controversial data about the potential role of TWEAK in different cardiovascular pathologies. NT-proBNP is an established biomarker of myocardial wall stress, associated with poor functional class in HCM. We hypothesized that effort capacity in patients with HCM could be related to serum levels of these biomarkers. MATERIALS AND METHODS We included 40 haemodynamic stable HCM patients and 53 healthy controls with similar sex and age. We studied exercise capacity by maximal oxygen consumption in a limited treadmill exercise test. TWEAK and NT-proBNP were assayed by ELISA method and automated Elecsys® platform, respectively. We obtained 46 samples of myocardial tissues by septal myectomy in patients with HCM and evaluated myocardial fibrosis, immunoreaction with TWEAK antibody and apoptosis with TUNEL assay. RESULTS We found raised TWEAK and NT-proBNP serum levels in patients when compared with control levels (both P < 0.001). In a multivariate analysis, TWEAK and NT-proBNP levels, as well as sex, remained independently associated with the effort capacity (all P < 0.05). We found an association between immunoreaction degree and the degree of myocardial fibrosis (P = 0.021), as well as apoptosis (P = 0.002) in the tissue samples from patients undergoing septal myectomy. CONCLUSIONS TWEAK and NT-proBNP levels are biomarkers of disease severity independently associated with the effort capacity in patients with HCM.
Collapse
Affiliation(s)
- Diana Hernández-Romero
- Hospital Universitario Virgen de la Arrixaca, Universidad de Murcia, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca (IMIB-Arrixaca), Murcia, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Padrão AI, Moreira-Gonçalves D, Oliveira PA, Teixeira C, Faustino-Rocha AI, Helguero L, Vitorino R, Santos LL, Amado F, Duarte JA, Ferreira R. Endurance training prevents TWEAK but not myostatin-mediated cardiac remodelling in cancer cachexia. Arch Biochem Biophys 2015; 567:13-21. [PMID: 25575785 DOI: 10.1016/j.abb.2014.12.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 12/09/2014] [Accepted: 12/29/2014] [Indexed: 12/15/2022]
Abstract
Strategies to prevent tumour burden-induced cardiac remodelling that might progress to heart failure are necessary to improve patients' health outcomes and tolerability to cancer therapies. Exercise has been suggested as a measure to prevent cardiac damage; however, its effectiveness on regulating cardiac remodelling secondary to cancer was never addressed. Using an animal model of mammary tumorigenesis, we studied the impact of 35weeks of endurance training on heart, focusing on the signalling pathways modulated by pro-inflammatory and wasting cytokines. The cardiac fibrosis and myofiber disorganization induced by tumour burden was paralleled by the increase of myostatin and TWEAK with the activation of signalling pathways involving Smad-3, NF-κB, TRAF-6 and atrogin-1. The activation of Akt/mTOR was observed in heart from rats with tumours, for which contributed the extracellular matrix. Endurance training prevented the increase of serum and cardiac TWEAK promoted by cancer, as well as the activation of NF-κB, TRAF6, atrogin-1 and p70S6K in heart. Data highlight the impact of exercise in the modulation of signalling pathways activated by wasting cytokines and the resulting outcomes on heart adaptation. Future studies focused on the cellular pathways underlying cardiac remodelling will assist in the development of exercise programs targeting cancer-related cardiac alterations.
Collapse
Affiliation(s)
- Ana Isabel Padrão
- QOPNA, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Daniel Moreira-Gonçalves
- CIAFEL, Faculty of Sport, University of Porto, Porto, Portugal; Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Paula A Oliveira
- CITAB, Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro, Vila Real, Portugal
| | - Catarina Teixeira
- CITAB, Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro, Vila Real, Portugal
| | - Ana I Faustino-Rocha
- CITAB, Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro, Vila Real, Portugal
| | - Luísa Helguero
- QOPNA, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Rui Vitorino
- QOPNA, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Porto, Portugal
| | - Francisco Amado
- QOPNA, School of Health Sciences, University of Aveiro, Portugal
| | | | - Rita Ferreira
- QOPNA, Department of Chemistry, University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
14
|
Yadava RS, Foff EP, Yu Q, Gladman JT, Kim YK, Bhatt KS, Thornton CA, Zheng TS, Mahadevan MS. TWEAK/Fn14, a pathway and novel therapeutic target in myotonic dystrophy. Hum Mol Genet 2014; 24:2035-48. [PMID: 25504044 DOI: 10.1093/hmg/ddu617] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1), the most prevalent muscular dystrophy in adults, is characterized by progressive muscle wasting and multi-systemic complications. DM1 is the prototype for disorders caused by RNA toxicity. Currently, no therapies exist. Here, we identify that fibroblast growth factor-inducible 14 (Fn14), a member of the tumor necrosis factor receptor super-family, is induced in skeletal muscles and hearts of mouse models of RNA toxicity and in tissues from DM1 patients, and that its expression correlates with severity of muscle pathology. This is associated with downstream signaling through the NF-κB pathways. In mice with RNA toxicity, genetic deletion of Fn14 results in reduced muscle pathology and better function. Importantly, blocking TWEAK/Fn14 signaling with an anti-TWEAK antibody likewise improves muscle histopathology and functional outcomes in affected mice. These results reveal new avenues for therapeutic development and provide proof of concept for a novel therapeutic target for which clinically available therapy exists to potentially treat muscular dystrophy in DM1.
Collapse
Affiliation(s)
| | - Erin P Foff
- Department of Neurology, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | | - Kirti S Bhatt
- Department of Neurology, University of Rochester, Rochester, NY 14642, USA and
| | - Charles A Thornton
- Department of Neurology, University of Rochester, Rochester, NY 14642, USA and
| | - Timothy S Zheng
- Department of Immunology, Biogen Idec, Cambridge, MA 02142, USA
| | | |
Collapse
|
15
|
Ates I, Ozkayar N, Akyel F, Topcuoglu C, Akyel S, Barça AN, Dede F. The relationship between asymptomatic organ damage, and serum soluble tumor necrosis factor-like weak inducer of apoptosis (sTWEAK) and Interleukin-17A (IL-17A) levels in non-diabetic hypertensive patients. BMC Nephrol 2014; 15:159. [PMID: 25273526 PMCID: PMC4190353 DOI: 10.1186/1471-2369-15-159] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 09/29/2014] [Indexed: 01/08/2023] Open
Abstract
Background This study aimed to measure the serum soluble tumor necrosis factor-like weak inducer of apoptosis (sTWEAK) and interleukin-17A (IL-17A) levels in hypertensive patients with/without asymptomatic organ damage (AOD), as well as to determine the relationship between the serum sTWEAK and IL17-A levels, and carotid intima media thickness (CIMT), proteinuria, retinopathy, and the left ventricle mass index (LVMI). Methods The study included 159 patients diagnosed with and followed-up for primary hypertension (HT); 79 of the patients had AOD (61 female and 18 male) and 80 did not (52 female and 28 male). sTWEAK and IL-17A levels were measured in all patients. Results The sTWEAK level was significantly lower in the patients with AOD than in those without AOD (858.4 pg/mL vs. 1151.58 pg/mL, P = 0.001). The sTWEAK level was negatively correlated with the mean microalbuminuria level and LVMI. The median IL-17A level was significantly higher in the patients with AOD than in those without AOD (2.34 pg/mL vs. 1.80 pg/mL, P = 0.001). There was a positive correlation between mean IL-17A level, and mean microalbuminuria level, CIMT, and LVMI. Multivariate logistic regression analysis showed that patient age, sTWEAK level, and mean 24-h systolic blood pressure were predictors of AOD. Conclusions The sTWEAK level was lower and IL-17A level was higher in the patients with AOD. It remains unknown if sTWEAK and IL-17A play a role in the pathophysiology of AOD. Prospective observational studies are needed to determine the precise role of sTWEAK and IL-17A in the development of target organ damage.
Collapse
Affiliation(s)
- Ihsan Ates
- Department of Nephrology, Ankara Numune Education and Research Hospital, Sıhhiye, Ankara 061100, Turkey.
| | | | | | | | | | | | | |
Collapse
|
16
|
Trebing J, Lang I, Chopra M, Salzmann S, Moshir M, Silence K, Riedel SS, Siegmund D, Beilhack A, Otto C, Wajant H. A novel llama antibody targeting Fn14 exhibits anti-metastatic activity in vivo. MAbs 2014; 6:297-308. [PMID: 24135629 PMCID: PMC3929451 DOI: 10.4161/mabs.26709] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 10/02/2013] [Accepted: 10/04/2013] [Indexed: 12/30/2022] Open
Abstract
Expression of fibroblast growth factor (FGF)-inducible 14 (Fn14), a member of the tumor necrosis factor receptor superfamily, is typically low in healthy adult organisms, but strong Fn14 expression is induced in tissue injury and tissue remodeling. High Fn14 expression is also observed in solid tumors, which is why this receptor is under consideration as a therapeutic target in oncology. Here, we describe various novel mouse-human cross-reactive llama-derived recombinant Fn14-specific antibodies (5B6, 18D1, 4G5) harboring the human IgG1 Fc domain. In contrast to recombinant variants of the established Fn14-specific antibodies PDL192 and P4A8, all three llama-derived antibodies efficiently bound to the W42A and R56P mutants of human Fn14. 18D1 and 4G5, but not 5B6, efficiently blocked TNF-like weak inducer of apoptosis(TWEA K) binding at low concentrations (0.2–2 μg/ml). Oligomerization and Fcγ receptor (FcγR) binding converted all antibodies into strong Fn14 agonists. Variants of 18D1 with enhanced and reduced antibody-dependent cell-mediated cytotoxicity (ADCC) activity were further analyzed in vivo with respect to their effect on metastasis. In a xenogeneic model using human colon carcinoma cancer cells, both antibody variants were effective in reducing metastasis to the liver. In contrast, only the 18D1 variant with enhanced ADCC activity, but not its ADCC-defective counterpart, suppressed lung metastasis in the RE NCA model. In sum, this suggests that Fn14 targeting might primarily act by triggering of antibody effector functions, but also by blockade of TWEA K-Fn14 interaction in some cases
Collapse
Affiliation(s)
- Johannes Trebing
- Division of Molecular Internal Medicine; Department of Internal Medicine II; University Hospital of Würzburg; Würzburg, Germany
| | - Isabell Lang
- Division of Molecular Internal Medicine; Department of Internal Medicine II; University Hospital of Würzburg; Würzburg, Germany
| | - Martin Chopra
- IZKF Research Laboratory for Experimental Stem Cell Transplantation; Department of Internal Medicine II; University Hospital of Würzburg; Würzburg, Germany
| | - Steffen Salzmann
- Division of Molecular Internal Medicine; Department of Internal Medicine II; University Hospital of Würzburg; Würzburg, Germany
| | | | | | - Simone S Riedel
- IZKF Research Laboratory for Experimental Stem Cell Transplantation; Department of Internal Medicine II; University Hospital of Würzburg; Würzburg, Germany
| | - Daniela Siegmund
- Division of Molecular Internal Medicine; Department of Internal Medicine II; University Hospital of Würzburg; Würzburg, Germany
| | - Andreas Beilhack
- IZKF Research Laboratory for Experimental Stem Cell Transplantation; Department of Internal Medicine II; University Hospital of Würzburg; Würzburg, Germany
| | - Christoph Otto
- Department of General, Visceral, Vascular, and Paediatric Surgery; University Hospital of Würzburg; Würzburg, Germany
| | - Harald Wajant
- Division of Molecular Internal Medicine; Department of Internal Medicine II; University Hospital of Würzburg; Würzburg, Germany
| |
Collapse
|
17
|
Gopal DM, Sam F. New and emerging biomarkers in left ventricular systolic dysfunction--insight into dilated cardiomyopathy. J Cardiovasc Transl Res 2013; 6:516-27. [PMID: 23609585 DOI: 10.1007/s12265-013-9462-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 03/27/2013] [Indexed: 01/16/2023]
Abstract
Dilated cardiomyopathy (DCM) is characterized by deteriorating cardiac performance, impaired contraction and dilation of the left ventricle (or both ventricles). Blood markers--known as "biomarkers"--allow insight into underlying pathophysiologic mechanisms and biologic pathways while predicting outcomes and guiding heart failure management and/or therapies. In this review, we provide an alternative approach to conceptualize heart failure biomarkers: the cardiomyocyte, its surrounding microenvironment, and the macroenvironment, integrating these entities which may impact cellular processes involved in the pathogenesis and/or propagation of DCM. Newer biomarkers of left ventricular systolic dysfunction can be categorized under: (a) myocyte stress and stretch, (b) myocyte apoptosis, (c) cardiac interstitium, (d) inflammation, (e) oxidative stress, (f) cardiac energetics, (g) neurohormones, and (h) renal biomarkers. Biomarkers provide insight into the pathogenesis of DCM while predicting and potentially providing prognostic information in these patients with heart failure.
Collapse
Affiliation(s)
- Deepa M Gopal
- Cardiovascular Section and Evans Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | | |
Collapse
|