1
|
Watany MM, Elhosary MM. Clinical utility of circulating TWEAK and CD163 as biomarkers of iron-induced cardiac decompensation in transfusion dependent thalassemia major. Cytokine 2024; 173:156443. [PMID: 38000169 DOI: 10.1016/j.cyto.2023.156443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/30/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023]
Abstract
BACKGROUND AND AIM Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) affects most of the cells involved in cardiac fibrosis like inflammatory cells, cardiomyocytes and fibroblasts. CD163, the receptor of TWEAK on the surface of type 2 macrophages, is shed into plasma upon macrophages activation. This work aimed to evaluate serum TWEAK and its decoy receptor CD163 as probable biomarkers to monitor myocardial iron overload (MIO) in transfusion dependent thalassemia major (TDTM) patients and to predict iron-induced cardiac decompensation (IICD). METHODS A total of 140 TDTM patients were enrolled. Patients were categorized into two groups; group I (n = 70) diagnosed with IICD while group II (n = 70) had no evidence of IICD. sTWEAK and sCD163 were quantitated utilizing Enzyme-linked-immunosorbent- assay. RESULTS sTWEAK was evidently lower in group I than group II (medians, 412 and 1052 pg/mL respectively). sCD163 was higher in group I than group II (medians, 615.5 and 323.5 ng/mL respectively). sTWEAK positively correlated with cardiac MRI-T2 mapping and ventricular ejection fractions and negatively correlated with B-Natriuretic peptide and cardiac troponin. An inverse relationship between TWEAK and CD163 was documented throughout the study. sTWEAK, sCD163 and TWEAK/CD163 ratio proved to be significant predictors of IICD in TDTM patients. TWEAK/CD163 ratio < 1.04 discriminated IICD in TDTM patients with 100 % clinical sensitivity and specificity. CONCLUSION Circulating TWEAK and CD163 appears to be promising biomarkers for monitoring MIO and predicting IICD in TDTM patients.
Collapse
Affiliation(s)
- Mona M Watany
- Clinical Pathology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt.
| | - Marwa M Elhosary
- Msc Immunology from Tanta University, Faculty of Science, Tanta 31527, Egypt
| |
Collapse
|
2
|
Chronic Trypanosoma cruzi infection activates the TWEAK/Fn14 axis in cardiac myocytes and fibroblasts driving structural and functional changes that affect the heart. Exp Parasitol 2023; 248:108491. [PMID: 36841467 DOI: 10.1016/j.exppara.2023.108491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 01/04/2023] [Accepted: 02/18/2023] [Indexed: 02/25/2023]
Abstract
Sustained interaction between the cytokine tumor necrosis factor-like weak inducer of apoptosis (TWEAK) and its functional receptor, fibroblast growth factor-inducible 14 (Fn14), has been linked to cardiovascular disorders. Chagas cardiomyopathy, elicited by Trypanosoma cruzi infection, is associated with chronic inflammation, fibrosis and hypertrophy. This study aimed to explore the involvement of the TWEAK/Fn 14 axis in development of Chagas heart disease. Parasite infection in vitro triggered Fn14 overexpression in atrial HL-1 myocytes and cardiac MCF fibroblasts. Fn14 levels were also increased in heart tissue from C57BL/6 mice at 130 days post-infection, particularly in myocytes and fibroblasts. Concurrently, TWEAK expression in circulating monocytes from this group was higher than that determined in uninfected controls. TWEAK/Fn14 interaction was functional in myocytes and fibroblasts isolated from infected hearts, leading to TNF receptor-associated factor 2 (TRAF2)-mediated activation of nuclear factor kappa B (NFκB) signaling. Ex vivo stimulation of both cell types with recombinant TWEAK for 24 h boosted the NFκB-regulated production of proinflammatory/profibrotic mediators (IL-1β, IL-6, TNF-α, IL-8, CCL2, CCL5, MMP-2, MMP-9, ICAM-1, E-selectin) involved in chronic T. cruzi cardiomyopathy. We further evaluated the therapeutic potential of the soluble decoy receptor Fn14-Fc to interfere with TWEAK/Fn14-dependent pathogenic activity. Fn14-Fc treatment of chronically infected mice was effective in neutralizing the ligand and reverting electrocardiographic abnormalities, maladaptive inflammation, adverse remodeling and hypertrophy in myocardium. Altogether, these findings suggest that sustained TWEAK/Fn14 induction by persistent T. cruzi infection is implicated in cardiopathogenesis and make TWEAK/Fn14 axis a promising target for the treatment of chronic Chagas heart disease.
Collapse
|
3
|
Zhang Y, Li X, Liu W, Hu G, Gu H, Cui X, Zhang D, Zeng W, Xia Y. TWEAK/Fn14 signaling may function as a reactive compensatory mechanism against extracellular matrix accumulation in keloid fibroblasts. Eur J Cell Biol 2023; 102:151290. [PMID: 36709605 DOI: 10.1016/j.ejcb.2023.151290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 01/22/2023] [Accepted: 01/22/2023] [Indexed: 01/25/2023] Open
Abstract
Overabundance of the extracellular matrix resulting from hyperproliferation of keloid fibroblasts (KFs) and dysregulation of apoptosis represents the main pathophysiology underlying keloids. TWEAK is a weak apoptosis inducer, and it plays a critical role in pathological tissue remodeling via its receptor, Fn14. However, the role of TWEAK/Fn14 signaling in the pathogenesis of keloids has not been investigated. In this study, we confirmed the overexpression levels of TWEAK and Fn14 in clinical keloid tissue specimens and primary KFs. The extracellular matrix (ECM)-related genes were also evaluated between primary KFs and their normal counterparts to determine the factors leading to the formation or development of keloids. Unexpectedly, exogenous TWEAK significantly reduced the levels of collagen I and collagen III, as well as alpha-smooth muscle actin (α-SMA). Additionally, TWEAK promoted MMPs expression and apoptosis activity of KFs. Furthermore, we verified that the inhibitory effect of TWEAK on KFs is through down-regulation of Polo-like kinase 5, which modulates cell differentiation and apoptosis. The TWEAK-Fn14 axis seems to be a secondary, although less effective, compensatory mechanism to increase the catabolic functions of fibroblasts in an attempt to further decrease the accumulation of collagen. DATA AVAILABILITY: All data generated or analyzed during this study are included in this published article (and its Supporting Information files).
Collapse
Affiliation(s)
- Yitian Zhang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Xiaoli Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004 China
| | - Wei Liu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Guanglei Hu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Hanjiang Gu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Xiao Cui
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Dewu Zhang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Weihui Zeng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| |
Collapse
|
4
|
Extracellular Vesicle-Associated TWEAK Contributes to Vascular Inflammation and Remodeling During Acute Cellular Rejection. JACC Basic Transl Sci 2023. [DOI: 10.1016/j.jacbts.2022.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
5
|
Osna NA, Rasineni K, Ganesan M, Donohue TM, Kharbanda KK. Pathogenesis of Alcohol-Associated Liver Disease. J Clin Exp Hepatol 2022; 12:1492-1513. [PMID: 36340300 PMCID: PMC9630031 DOI: 10.1016/j.jceh.2022.05.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022] Open
Abstract
Excessive alcohol consumption is a global healthcare problem with enormous social, economic, and clinical consequences. While chronic, heavy alcohol consumption causes structural damage and/or disrupts normal organ function in virtually every tissue of the body, the liver sustains the greatest damage. This is primarily because the liver is the first to see alcohol absorbed from the gastrointestinal tract via the portal circulation and second, because the liver is the principal site of ethanol metabolism. Alcohol-induced damage remains one of the most prevalent disorders of the liver and a leading cause of death or transplantation from liver disease. Despite extensive research on the pathophysiology of this disease, there are still no targeted therapies available. Given the multifactorial mechanisms for alcohol-associated liver disease pathogenesis, it is conceivable that a multitherapeutic regimen is needed to treat different stages in the spectrum of this disease.
Collapse
Key Words
- AA, Arachidonic acid
- ADH, Alcohol dehydrogenase
- AH, Alcoholic hepatitis
- ALD, Alcohol-associated liver disease
- ALDH, Aldehyde dehydrogenase
- ALT, Alanine transaminase
- ASH, Alcohol-associated steatohepatitis
- AST, Aspartate transaminase
- AUD, Alcohol use disorder
- BHMT, Betaine-homocysteine-methyltransferase
- CD, Cluster of differentiation
- COX, Cycloxygenase
- CTLs, Cytotoxic T-lymphocytes
- CYP, Cytochrome P450
- CYP2E1, Cytochrome P450 2E1
- Cu/Zn SOD, Copper/zinc superoxide dismutase
- DAMPs, Damage-associated molecular patterns
- DC, Dendritic cells
- EDN1, Endothelin 1
- ER, Endoplasmic reticulum
- ETOH, Ethanol
- EVs, Extracellular vesicles
- FABP4, Fatty acid-binding protein 4
- FAF2, Fas-associated factor family member 2
- FMT, Fecal microbiota transplant
- Fn14, Fibroblast growth factor-inducible 14
- GHS-R1a, Growth hormone secretagogue receptor type 1a
- GI, GOsteopontinastrointestinal tract
- GSH Px, Glutathione peroxidase
- GSSG Rdx, Glutathione reductase
- GST, Glutathione-S-transferase
- GWAS, Genome-wide association studies
- H2O2, Hydrogen peroxide
- HA, Hyaluronan
- HCC, Hepatocellular carcinoma
- HNE, 4-hydroxynonenal
- HPMA, 3-hydroxypropylmercapturic acid
- HSC, Hepatic stellate cells
- HSD17B13, 17 beta hydroxy steroid dehydrogenase 13
- HSP 90, Heat shock protein 90
- IFN, Interferon
- IL, Interleukin
- IRF3, Interferon regulatory factor 3
- JAK, Janus kinase
- KC, Kupffer cells
- LCN2, Lipocalin 2
- M-D, Mallory–Denk
- MAA, Malondialdehyde-acetaldehyde protein adducts
- MAT, Methionine adenosyltransferase
- MCP, Macrophage chemotactic protein
- MDA, Malondialdehyde
- MIF, Macrophage migration inhibitory factor
- Mn SOD, Manganese superoxide dismutase
- Mt, Mitochondrial
- NK, Natural killer
- NKT, Natural killer T-lymphocytes
- OPN, Osteopontin
- PAMP, Pathogen-associated molecular patterns
- PNPLA3, Patatin-like phospholipase domain containing 3
- PUFA, Polyunsaturated fatty acid
- RIG1, Retinoic acid inducible gene 1
- SAH, S-adenosylhomocysteine
- SAM, S-adenosylmethionine
- SCD, Stearoyl-CoA desaturase
- STAT, Signal transduction and activator of transcription
- TIMP1, Tissue inhibitor matrix metalloproteinase 1
- TLR, Toll-like receptor
- TNF, Tumor necrosis factor-α
- alcohol
- alcohol-associated liver disease
- ethanol metabolism
- liver
- miRNA, MicroRNA
- p90RSK, 90 kDa ribosomal S6 kinase
Collapse
Affiliation(s)
- Natalia A. Osna
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
- Department of Internal Medicine, Omaha, NE, 68198, USA
| | - Karuna Rasineni
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
- Department of Internal Medicine, Omaha, NE, 68198, USA
| | - Murali Ganesan
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
- Department of Internal Medicine, Omaha, NE, 68198, USA
| | - Terrence M. Donohue
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
- Department of Internal Medicine, Omaha, NE, 68198, USA
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kusum K. Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
- Department of Internal Medicine, Omaha, NE, 68198, USA
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
6
|
Li R, Wang TY, Shelp-Peck E, Wu SP, DeMayo FJ. The single-cell atlas of cultured human endometrial stromal cells. F&S SCIENCE 2022; 3:349-366. [PMID: 36089208 PMCID: PMC9669198 DOI: 10.1016/j.xfss.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To systematically analyze the cell composition and transcriptome of primary human endometrial stromal cells (HESCs) and transformed human endometrial stromal cells (THESCs). DESIGN The primary HESCs from 3 different donors and 1 immortalized THESC were collected from the human endometrium at the midsecretory phase and cultured in vitro. SETTING Academic research laboratory. PATIENT(S) None. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Single-cell ribonucleic acid sequencing analysis. RESULT(S) We found the individual differences among the primary HESCs and bigger changes between the primary HESCs and THESCs. Cell clustering with or without integration identified cell clusters belonging to mature, proliferative, and active fibroblasts that were conserved across all samples at different stages of the cell cycles with intensive cell communication signals. All primary HESCs and THESCs can be correlated with some subpopulations of fibroblasts in the human endometrium. CONCLUSION(S) Our study indicated that the primary HESCs and THESCs displayed conserved cell characters and distinct cell clusters. Mature, proliferative, and active fibroblasts at different stages or cell cycles were detected across all samples and presented with a complex cell communication network. The cultured HESCs and THESCs retained the features of some subpopulations within the human endometrium.
Collapse
Affiliation(s)
- Rong Li
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina
| | - Tian-Yuan Wang
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina
| | - Elinor Shelp-Peck
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina; The Biological Sciences Department, The Department of Chemistry, Physics, and Geosciences, Meredith College, Raleigh, North Carolina
| | - San-Pin Wu
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina
| | - Francesco J DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina.
| |
Collapse
|
7
|
Ray S, Adelnia H, Ta HT. Collagen and the effect of poly-l-lactic acid based materials on its synthesis. Biomater Sci 2021; 9:5714-5731. [PMID: 34296717 DOI: 10.1039/d1bm00516b] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Collagen is an important protein in various biological functions such as providing elasticity and waterproofing to the skin, structural stability to the cells in connective tissues (e.g. tendons, and bone) and stabilisation of atherosclerotic plaques. Collagen as a peptide with a peculiar triple helical structure is majorly composed of glycine and proline amino acids and is synthesised by fibroblasts via intracellular and extracellular mechanisms. Collagen plays an important role in wound healing, bone repair and plaque build-up during atherosclerosis. Various factors such as interleukins, insulin-like growth factor-I, nicotine, and glucose have been shown to influence collagen synthesis. This paper provides an overview of collagen structure, synthesis mechanisms, and the parameters that stimulate those mechanisms. Poly-l-lactic acid as a well-known biocompatible and biodegradable polymer has proved to stimulate collagen synthesis in various physical forms. As such, in this review special emphasis is laid on the effects of poly-l-lactic acid as well as its mechanism of action on collagen synthesis.
Collapse
Affiliation(s)
- Subarna Ray
- Queensland Micro- and Nanotechnology Centre, Griffith University, Brisbane, 4111, Queensland, Australia.
| | | | | |
Collapse
|
8
|
Empagliflozin Disrupts a Tnfrsf12a-Mediated Feed Forward Loop That Promotes Left Ventricular Hypertrophy. Cardiovasc Drugs Ther 2021; 36:619-632. [PMID: 33886003 DOI: 10.1007/s10557-021-07190-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/14/2021] [Indexed: 01/10/2023]
Abstract
PURPOSE Although the cardioprotective benefits of sodium-glucose cotransporter 2 (SGLT2) inhibitors are now widely appreciated, the mechanisms underlying these benefits remain unresolved. Tumor necrosis factor receptor superfamily member 12a (Tnfrsf12a) is a receptor for tumor necrosis factor superfamily member 12 (Tnfsf12). Tnfrsf12a is highly inducible and plays a key role in the development of cardiac hypertrophy and heart failure. Here we set out to determine if SGLT2 inhibition affects the Tnfsf12/Tnfrsf12a system in the stressed myocardium. METHODS C57BL/6N mice that had undergone sham or transverse aortic constriction (TAC) surgery were treated with either the SGLT2 inhibitor empagliflozin (400 mg/kg diet; 60-65 mg/kg/day) or standard chow alone and were followed for 8 weeks. Tnfrsf12a expression in mouse hearts was assessed by in situ hybridization, qRT-PCR, and immunoblotting. RESULTS Left ventricular (LV) mass, end-systolic volume, and end-diastolic volume were all increased in TAC mice and were significantly lower with empagliflozin. Myocyte hypertrophy and interstitial fibrosis in TAC hearts were similarly attenuated with empagliflozin. Tnfrsf12a expression was upregulated in mouse hearts following TAC surgery but not in the hearts of empagliflozin-treated mice. In cultured cardiomyocytes, Tnfrsf12a antagonism attenuated the increase in cardiomyocyte size that was induced by phenylephrine. CONCLUSION Empagliflozin attenuates LV enlargement in mice with hypertrophic heart failure. This effect may be mediated, at least in part, by a reduction in loading conditions which limits upregulation of the inducible, proinflammatory, and prohypertrophic TNF superfamily receptor, Tnfrsf12a. Disruption of the Tnfsf12/Tnfrsf12a feed forward system may contribute to the cardioprotective benefits of SGLT2 inhibition.
Collapse
|
9
|
Poveda J, Vázquez-Sánchez S, Sanz AB, Ortiz A, Ruilope LM, Ruiz-Hurtado G. TWEAK-Fn14 as a common pathway in the heart and the kidneys in cardiorenal syndrome. J Pathol 2021; 254:5-19. [PMID: 33512736 DOI: 10.1002/path.5631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/23/2020] [Accepted: 01/12/2021] [Indexed: 12/19/2022]
Abstract
There is a complex relationship between cardiac and renal disease, often referred to as the cardiorenal syndrome. Heart failure adversely affects kidney function, and both acute and chronic kidney disease are associated with structural and functional changes to the myocardium. The pathological mechanisms and contributing interactions that surround this relationship remain poorly understood, limiting the opportunities for therapeutic intervention. The cytokine tumor necrosis factor-like weak inducer of apoptosis (TWEAK) and its receptor, fibroblast growth factor-inducible 14 (Fn14), are abundantly expressed in injured kidneys and heart. The TWEAK-Fn14 axis promotes responses that drive tissue injury such as inflammation, proliferation, fibrosis, and apoptosis, while restraining the expression of tissue protective factors such as the anti-aging factor Klotho and the master regulator of mitochondrial biogenesis peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α). High levels of TWEAK induce cardiac remodeling, and promote inflammation, tubular and podocyte injury and death, fibroblast proliferation, and, ultimately, renal fibrosis. Accordingly, targeting the TWEAK-Fn14 axis is protective in experimental kidney and heart disease. TWEAK has also emerged as a biomarker of kidney damage and cardiovascular outcomes and has been successfully targeted in clinical trials. In this review, we update our current knowledge of the roles of the TWEAK-Fn14 axis in cardiovascular and kidney disease and its potential contribution to the cardiorenal syndrome. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jonay Poveda
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Sara Vázquez-Sánchez
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Ana B Sanz
- Research Institute - Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain.,REDINREN, Madrid, Spain
| | - Alberto Ortiz
- Research Institute - Fundacion Jimenez Diaz, Autonoma University, Madrid, Spain.,REDINREN, Madrid, Spain
| | - Luis M Ruilope
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain.,School of Doctoral Studies and Research, European University of Madrid, Madrid, Spain.,CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Gema Ruiz-Hurtado
- Cardiorenal Translational Laboratory, Institute of Research i+12, Hospital Universitario 12 de Octubre, Madrid, Spain.,CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain
| |
Collapse
|
10
|
Zhang Y, Zeng W, Xia Y. TWEAK/Fn14 axis is an important player in fibrosis. J Cell Physiol 2020; 236:3304-3316. [PMID: 33000480 DOI: 10.1002/jcp.30089] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/15/2022]
Abstract
Fibrosis is a common pathological condition associated with abnormal repair after tissue injury. However, the etiology and molecular mechanisms of fibrosis are still not well-understood. Tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) belongs to the TNF superfamily and acts by binding to its receptor, fibroblast growth factor-inducible 14 (Fn14), thereby activating a variety of intracellular signal transduction pathways in various types of cells. Besides promoting the expression of growth factors, activation of TWEAK/Fn14 signaling after tissue injury can promote the expression of pro-inflammatory cytokines, which trigger the immune response, thereby exacerbating the injury. Severe or repetitive injury leads to a dysregulated tissue repair process, in which the TWEAK/Fn14 axis promotes the activation and proliferation of myofibroblasts, induces the secretion of the extracellular matrix, and regulates profibrotic mediators to further perpetuate and sustain the fibrotic process. In this review, we summarize the available experimental evidence on the underlying molecular mechanisms by which the TWEAK/Fn14 pathway mediates the development and progression of fibrosis. In addition, we discuss the therapeutic potential of the TWEAK/Fn14 pathway in fibrosis-associated diseases based on evidence derived from multiple models and cells from injured tissue and fibrotic tissue.
Collapse
Affiliation(s)
- Yitian Zhang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Weihui Zeng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
11
|
Wang M, Xie Z, Xu J, Feng Z. TWEAK/Fn14 axis in respiratory diseases. Clin Chim Acta 2020; 509:139-148. [PMID: 32526219 DOI: 10.1016/j.cca.2020.06.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/31/2020] [Accepted: 06/03/2020] [Indexed: 02/08/2023]
Abstract
Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is a well known multifunctional cytokine extensively distributed in cell types and tissues. Accumulating evidence has shown that TWEAK binding to the receptor factor-inducible 14 (Fn14) participates in diverse pathologic processes including cell proliferation and death, angiogenesis, carcinogenesis and inflammation. Interestingly, alterations of intracellular signaling cascades are correlated to the development of respiratory disease. Recently, a several lines of evidence suggests that TWEAK in lung tissues are closely associated with these signaling pathways. In this review, we explore if TWEAK could provide a novel therapeutic strategy for managing respiratory disease in general and pulmonary arterial hypertension (PAH), obstructive sleep apnea syndrome (OSAS), asthma, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and non-small cell lung cancer (NSCLC), specifically.
Collapse
Affiliation(s)
- Min Wang
- Department of Otorhinolaryngology, University of South China Affiliated Nanhua Hospital, Hengyang 421002, China
| | - Zhijuan Xie
- Department of Nephrology, The First Affiliated Hospital of University of South China, Hengyang 421001, China
| | - Jin Xu
- School of Pharmaceutical Sciences, Changsha Medical University, Changsha 410219, Hunan, China.
| | - Zhuyu Feng
- Department of Critical Care Medicine, University of South China Affiliated Nanhua Hospital, Hengyang 421002, China.
| |
Collapse
|
12
|
Stienen S, Ferreira JP, Kobayashi M, Preud'homme G, Dobre D, Machu JL, Duarte K, Bresso E, Devignes MD, López N, Girerd N, Aakhus S, Ambrosio G, Brunner-La Rocca HP, Fontes-Carvalho R, Fraser AG, van Heerebeek L, Heymans S, de Keulenaer G, Marino P, McDonald K, Mebazaa A, Papp Z, Raddino R, Tschöpe C, Paulus WJ, Zannad F, Rossignol P. Enhanced clinical phenotyping by mechanistic bioprofiling in heart failure with preserved ejection fraction: insights from the MEDIA-DHF study (The Metabolic Road to Diastolic Heart Failure). Biomarkers 2020; 25:201-211. [PMID: 32063068 DOI: 10.1080/1354750x.2020.1727015] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background: Heart failure with preserved ejection fraction (HFpEF) is a heterogeneous syndrome for which clear evidence of effective therapies is lacking. Understanding which factors determine this heterogeneity may be helped by better phenotyping. An unsupervised statistical approach applied to a large set of biomarkers may identify distinct HFpEF phenotypes.Methods: Relevant proteomic biomarkers were analyzed in 392 HFpEF patients included in Metabolic Road to Diastolic HF (MEDIA-DHF). We performed an unsupervised cluster analysis to define distinct phenotypes. Cluster characteristics were explored with logistic regression. The association between clusters and 1-year cardiovascular (CV) death and/or CV hospitalization was studied using Cox regression.Results: Based on 415 biomarkers, we identified 2 distinct clusters. Clinical variables associated with cluster 2 were diabetes, impaired renal function, loop diuretics and/or betablockers. In addition, 17 biomarkers were higher expressed in cluster 2 vs. 1. Patients in cluster 2 vs. those in 1 experienced higher rates of CV death/CV hospitalization (adj. HR 1.93, 95% CI 1.12-3.32, p = 0.017). Complex-network analyses linked these biomarkers to immune system activation, signal transduction cascades, cell interactions and metabolism.Conclusion: Unsupervised machine-learning algorithms applied to a wide range of biomarkers identified 2 HFpEF clusters with different CV phenotypes and outcomes. The identified pathways may provide a basis for future research.Clinical significanceMore insight is obtained in the mechanisms related to poor outcome in HFpEF patients since it was demonstrated that biomarkers associated with the high-risk cluster were related to the immune system, signal transduction cascades, cell interactions and metabolismBiomarkers (and pathways) identified in this study may help select high-risk HFpEF patients which could be helpful for the inclusion/exclusion of patients in future trials.Our findings may be the basis of investigating therapies specifically targeting these pathways and the potential use of corresponding markers potentially identifying patients with distinct mechanistic bioprofiles most likely to respond to the selected mechanistically targeted therapies.
Collapse
Affiliation(s)
- Susan Stienen
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - João Pedro Ferreira
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France.,Department of Physiology and Cardiothoracic Surgery, Cardiovascular Research and Development Unit, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Masatake Kobayashi
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - Gregoire Preud'homme
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - Daniela Dobre
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France.,Clinical research and Investigation Unit, Psychotherapeutic Center of Nancy, Laxou, France
| | - Jean-Loup Machu
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - Kevin Duarte
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - Emmanuel Bresso
- Equipe CAPSID, LORIA (CNRS, Inria NGE, Université de Lorraine), Vandoeuvre-lès-Nancy, France
| | | | - Natalia López
- Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Nicolas Girerd
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - Svend Aakhus
- Department of Cardiology and Institute for Surgical Research, Oslo University Hospital, Oslo, Norway.,ISB, Norwegian University of Science and Technology, Trondheim, Norway
| | - Giuseppe Ambrosio
- Division of Cardiology, University of Perugia School of Medicine, Perugia, Italy
| | | | - Ricardo Fontes-Carvalho
- Department of Surgery and Physiology, Cardiovascular Research Unit (UnIC), Faculty of Medicine, University of Porto, Porto, Portugal
| | - Alan G Fraser
- Wales Heart Research Institute, Cardiff University, Cardiff, UK
| | - Loek van Heerebeek
- Department of Cardiology, Onze Lieve Vrouwe Gasthuis, Amsterdam, the Netherlands
| | - Stephane Heymans
- Department of Cardiology, CARIM School for Cardiovascular Diseases Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands.,Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, Leuven, Belgium.,William Harvey Research Institute, Barts Heart Centre, Queen Mary University of London, London, UK
| | - Gilles de Keulenaer
- Laboratory of Physiopharmacology, Antwerp University, and ZNA Hartcentrum, Antwerp, Belgium
| | - Paolo Marino
- Clinical Cardiology, Università del Piemonte Orientale, Department of Translational Medicine, Azienda Ospedaliero Universitaria "Maggiore della Carità", Novara, Italy
| | - Kenneth McDonald
- School of Medicine and Medical Sciences, St Michael's Hospital Dun Laoghaire Co. Dublin, Dublin, Ireland
| | - Alexandre Mebazaa
- Department of Anaesthesiology and Critical Care Medicine, Saint Louis and Lariboisière University Hospitals and INSERM UMR-S 942, Paris, France
| | - Zoltàn Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Riccardo Raddino
- Department of Cardiology, Spedali Civili di Brescia, Brescia, Italy
| | - Carsten Tschöpe
- Department of Cardiology, Campus Virchow-Klinikum, C, Harite Universitaetsmedizin Berlin, Berlin Institute of Health - Center for Regenerative Therapies (BIH-BCRT), and the German Center for Cardiovascular Research (DZHK; Berlin partner site), Berlin, Germany
| | - Walter J Paulus
- Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Faiez Zannad
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| | - Patrick Rossignol
- CHRU de Nancy, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), INSERM U1116, Centre d'Investigation Clinique et Plurithématique 1433, INSERM, Université de Lorraine, Nancy, France
| |
Collapse
|
13
|
Unudurthi SD, Nassal DM, Patel NJ, Thomas E, Yu J, Pierson CG, Bansal SS, Mohler PJ, Hund TJ. Fibroblast growth factor-inducible 14 mediates macrophage infiltration in heart to promote pressure overload-induced cardiac dysfunction. Life Sci 2020; 247:117440. [PMID: 32070706 DOI: 10.1016/j.lfs.2020.117440] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/07/2020] [Accepted: 02/14/2020] [Indexed: 12/18/2022]
Abstract
AIMS Heart failure (HF) is characterized by compromised cardiac structure and function. Previous work has identified a link between upregulation of pro-inflammatory cytokines and HF. Tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) is a pro-inflammatory cytokine, which binds to fibroblast growth factor inducible 14 (Fn14), a ubiquitously expressed cell-surface receptor. The objective of this study was to investigate the role of TWEAK/Fn14 pathway in promoting cardiac inflammation under non ischemic stress conditions. MAIN METHODS Wild type (WT) and Fn14 knock out (Fn14-/-) mice were subjected to pressure overload [transaortic constriction (TAC)] for 1 or 6 weeks. A subset of WT TAC animals were treated with the Fn14 antagonist L524-0366. Cardiac function was measured by echocardiography. Cardiac fibrosis and macrophage infiltration were quantified using immunohistochemistry and flow cytometry, respectively. Cardiac fibroblasts were isolated for quantifying TWEAK-induced chemokine release. KEY FINDINGS Fn14-/- mice displayed improved cardiac function, reduced fibrosis and lower macrophage infiltration in heart compared to WT following TAC. L524-0366 mitigated maladaptive remodeling with TAC. TWEAK induced secretion of the pro-inflammatory chemokine, monocyte chemoattractant protein 1 from WT but not Fn14-/- fibroblasts in vitro, in part through activation of non-canonical NF-κB signaling. Finally, Fn14 expression was increased in mouse following TAC and in human failing hearts. SIGNIFICANCE Our findings support an important role for the TWEAK/Fn14 promoting macrophage infiltration and fibrosis in heart under non-ischemic stress, with potential for therapeutic intervention to improve cardiac function in the setting of HF.
Collapse
Affiliation(s)
- Sathya D Unudurthi
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA.
| | - Drew M Nassal
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Nehal J Patel
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Evelyn Thomas
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Jane Yu
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Curtis G Pierson
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Shyam S Bansal
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology & Cell Biology, USA
| | - Peter J Mohler
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology & Cell Biology, USA; Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Thomas J Hund
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, USA; Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
14
|
Tumor Necrosis Factor-Like Weak Inducer of Apoptosis (TWEAK)/Fibroblast Growth Factor-Inducible 14 (Fn14) Axis in Cardiovascular Diseases: Progress and Challenges. Cells 2020; 9:cells9020405. [PMID: 32053869 PMCID: PMC7072601 DOI: 10.3390/cells9020405] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVD) are the leading cause of mortality in Western countries. CVD include several pathologies, such as coronary artery disease, stroke, peripheral artery disease, and aortic aneurysm, among others. All of them are characterized by a pathological vascular remodeling in which inflammation plays a key role. Interaction between different members of the tumor necrosis factor superfamily and their cognate receptors induce several biological actions that may participate in CVD. The cytokine tumor necrosis factor-like weak inducer of apoptosis (TWEAK) and its functional receptor, fibroblast growth factor-inducible 14 (Fn14), are abundantly expressed during pathological cardiovascular remodeling. The TWEAK/Fn14 axis controls a variety of cellular functions, such as proliferation, differentiation, and apoptosis, and has several biological functions, such as inflammation and fibrosis that are linked to CVD. It has been demonstrated that persistent TWEAK/Fn14 activation is involved in both vessel and heart remodeling associated with acute and chronic CVD. In this review, we summarized the role of the TWEAK/Fn14 axis during pathological cardiovascular remodeling, highlighting the cellular components and the signaling pathways that are involved in these processes.
Collapse
|
15
|
Li C, Wang P, Fu Z, Li Y, Li S. Swainsonine inhibits proliferation and collagen synthesis of NIH-3T3 cells by declining miR-21. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2256-2264. [PMID: 31184213 DOI: 10.1080/21691401.2019.1620255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Chao Li
- Department of Burn and Plastic Surgery, Heze Municipal Hospital, Heze, China
| | - Peipei Wang
- Department of Burn and Plastic Surgery, Heze Municipal Hospital, Heze, China
| | - Ziyang Fu
- Department of Burn and Plastic Surgery, Heze Municipal Hospital, Heze, China
| | - Yongtao Li
- Department of Burn and Plastic Surgery, Heze Municipal Hospital, Heze, China
| | - Shouju Li
- Department of Burn and Plastic Surgery, Heze Municipal Hospital, Heze, China
| |
Collapse
|
16
|
Active Constituent in the Ethyl Acetate Extract Fraction of Terminalia bellirica Fruit Exhibits Antioxidation, Antifibrosis, and Proapoptosis Capabilities In Vitro. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5176090. [PMID: 31210842 PMCID: PMC6532289 DOI: 10.1155/2019/5176090] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/14/2019] [Accepted: 04/21/2019] [Indexed: 02/06/2023]
Abstract
Terminalia bellirica (Gaertn.) Roxb. fruit (TBF) is a widely planted traditional medicinal herb in Tibet. We aimed to determine the most active substance-enriched extract by comparing the in vitro antioxidant activities of different extract fractions of TBF that were subsequently extracted by petroleum ether, chloroform, ethyl acetate, and n-butanol after initial extraction by 95% ethanol. The main compounds of the ethyl acetate extract fraction (EF) were analyzed via HPLC-MS. Gallic acid (GA) was obtained from EF to determine in vitro antifibrotic activity based on the traditional usage of TBF. After HSC-T6 cells were incubated with GA, extracellular secreted levels of fibrosis-associated cytokines, such as collagen I, collagen III, TGF-β1, and hydroxyproline, were estimated by ELISA. Gene and protein expressions of PDGFR, CTGF, NF-κB, MMP-2, TIMP-1, TIMP-2, α-SMA, and the Bcl-2/Bax family were determined by quantitative PCR and western blot. The proapoptotic effect of GA was further investigated by annexin V-PI and TUNEL staining. These results indicate that EF has prominent in vitro antioxidant activity among four extract fractions, and its main component, GA, manifests antifibrosis activity and its potential mechanism of action includes inhibition of cytokine secretion and collagen synthesis, as well as proapoptosis of HSCs.
Collapse
|
17
|
Pedro Ferreira J, Verdonschot J, Collier T, Wang P, Pizard A, Bär C, Björkman J, Boccanelli A, Butler J, Clark A, Cleland JG, Delles C, Diez J, Girerd N, González A, Hazebroek M, Huby AC, Jukema W, Latini R, Leenders J, Levy D, Mebazaa A, Mischak H, Pinet F, Rossignol P, Sattar N, Sever P, Staessen JA, Thum T, Vodovar N, Zhang ZY, Heymans S, Zannad F. Proteomic Bioprofiles and Mechanistic Pathways of Progression to Heart Failure. Circ Heart Fail 2019; 12:e005897. [PMID: 31104495 PMCID: PMC8361846 DOI: 10.1161/circheartfailure.118.005897] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/16/2019] [Indexed: 02/06/2023]
Abstract
Background Identifying the mechanistic pathways potentially associated with incident heart failure (HF) may provide a basis for novel preventive strategies. Methods and Results To identify proteomic biomarkers and the potential underlying mechanistic pathways that may be associated with incident HF defined as the first hospitalization for HF, a nested-matched case-control design was used with cases (incident HF) and controls (without HF) selected from 3 cohorts (>20 000 individuals). Controls were matched on cohort, follow-up time, age, and sex. Two independent sample sets (a discovery set with 286 cases and 591 controls and a replication set with 276 cases and 280 controls) were used to discover and replicate the findings. Two hundred fifty-two circulating proteins in the plasma were studied. Adjusting for the matching variables age, sex, and follow-up time (and correcting for multiplicity of tests), 89 proteins were found to be associated with incident HF in the discovery phase, of which 38 were also associated with incident HF in the replication phase. These 38 proteins pointed to 4 main network clusters underlying incident HF: (1) inflammation and apoptosis, indicated by the expression of the TNF (tumor necrosis factor)-family members; (2) extracellular matrix remodeling, angiogenesis and growth, indicated by the expression of proteins associated with collagen metabolism, endothelial function, and vascular homeostasis; (3) blood pressure regulation, indicated by the expression of natriuretic peptides and proteins related to the renin-angiotensin-aldosterone system; and (4) metabolism, associated with cholesterol and atherosclerosis. Conclusions Clusters of biomarkers associated with mechanistic pathways leading to HF were identified linking inflammation, apoptosis, vascular function, matrix remodeling, blood pressure control, and metabolism. These findings provide important insight on the pathophysiological mechanisms leading to HF. Clinical Trial Registration: URL: https://www.clinicaltrials.gov . Unique identifier: NCT02556450.
Collapse
Affiliation(s)
- João Pedro Ferreira
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques-Plurithématique 14–33, and Inserm U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France, Department of Physiology and Cardiothoracic Surgery, Cardiovascular Research and Development Unit, Faculty of Medicine, University of Porto, Portugal
| | - Job Verdonschot
- Department of Cardiology, Maastricht University Medical Centre, Center for Heart Failure Research, Cardiovascular Research Institute Maastricht (CARIM), University Hospital Maastricht, the Netherlands, Department of Clinical Genetics, Maastricht University Medical Center, the Netherlands
| | - Timothy Collier
- London School of Hygiene and Tropical Medicine, United Kingdom
| | - Ping Wang
- Department of Clinical Genetics, Maastricht University Medical Center, the Netherlands
| | - Anne Pizard
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques-Plurithématique 14–33, and Inserm U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France, Inserm 1024, Institut de Biologie de l’École Normale Supérieure (IBENS), PSL University of Paris, France
| | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Germany
| | - Jens Björkman
- Department of Medicine, University of Mississippi School of Medicine, Jackson, Excellence Cluster REBIRTH, Hannover Medical School, Germany
| | | | | | - Andrew Clark
- Hull York Medical School, Castle Hill Hospital, Cottingham, United Kingdom
| | - John G. Cleland
- Robertson Centre for Biostatistics and Clinical Trials, Institute of Health and Wellbeing, Glasgow, United Kingdom, National Heart and Lung Institute, Royal Brompton and Harefield Hospitals, Imperial College, University of Glasgow, London, United Kingdom
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Scotland, United Kingdom
| | - Javier Diez
- Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain, CIBERCV, Carlos III Institute of Health, Madrid, Spain, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Spain, Departments of Nephrology, and Cardiology and Cardiac Surgery, University of Navarra Clinic, Pamplona, Spain
| | - Nicolas Girerd
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques-Plurithématique 14–33, and Inserm U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
| | - Arantxa González
- Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain, CIBERCV, Carlos III Institute of Health, Madrid, Spain, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Spain
| | - Mark Hazebroek
- Department of Cardiology, Maastricht University Medical Centre, Center for Heart Failure Research, Cardiovascular Research Institute Maastricht (CARIM), University Hospital Maastricht, the Netherlands
| | - Anne-Cécile Huby
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques-Plurithématique 14–33, and Inserm U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
| | - Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, the Netherlands
| | - Roberto Latini
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy
| | | | - Daniel Levy
- National Heart, Lung, and Blood Institute’s and Boston University’s Framingham Heart Study, MA, Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Alexandre Mebazaa
- UMRS 942, University Paris Diderot; APHP, University Hospitals Saint Louis Lariboisière, France
| | | | - Florence Pinet
- Inserm U1167, Institut Pasteur de Lille, Université de Lille, FHU-REMOD-VHF, France
| | - Patrick Rossignol
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques-Plurithématique 14–33, and Inserm U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | - Peter Sever
- International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London, England
| | - Jan A. Staessen
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Belgium, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, the Netherlands
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Germany, National Heart and Lung Institute, Imperial College London, United Kingdom
| | - Nicolas Vodovar
- UMRS 942, University Paris Diderot; APHP, University Hospitals Saint Louis Lariboisière, France
| | - Zhen-Yu Zhang
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Belgium
| | - Stephane Heymans
- Department of Cardiology, Maastricht University Medical Centre, Center for Heart Failure Research, Cardiovascular Research Institute Maastricht (CARIM), University Hospital Maastricht, the Netherlands, Department of Cardiovascular Research, University of Leuven, Belgium, Netherlands Heart Institute (ICIN), Utrecht, the Netherlands
| | - Faiez Zannad
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques-Plurithématique 14–33, and Inserm U1116, CHRU, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
| |
Collapse
|
18
|
Sheng Z, Ju C, Li B, Chen Z, Pan X, Yan G, He Y, Yao Y, Ma G. TWEAK promotes endothelial progenitor cell vasculogenesis to alleviate acute myocardial infarction via the Fn14-NF-κB signaling pathway. Exp Ther Med 2018; 16:4019-4029. [PMID: 30344680 PMCID: PMC6176210 DOI: 10.3892/etm.2018.6703] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 05/25/2018] [Indexed: 12/11/2022] Open
Abstract
Acute myocardial infarction (AMI) remains one of the leading causes of mortality worldwide; however, endothelial progenitor cell (EPC) transplantation has been proposed as a promising treatment strategy for EPC. High levels of tumor necrosis factor-related weak inducer of apoptosis (TWEAK) have been reported in AMI, although its effect on EPCs has not been reported. In the present study, immunofluorescence and flow cytometry were performed to assess the effect of TWEAK in isolated mouse EPCs. Echocardiography was used to evaluate the cardiac function of murine hearts following EPC treatment in the AMI model, while collagen synthesis within the heart tissue was assessed using Masson's trichrome staining. A tube formation assay and Transwell migration assay were performed to investigate the effects of TWEAK on vessel formation and EPC migration in vitro. Angiogenesis and arteriogenesis were assessed in vivo using immunohistochemistry and western blotting was performed to determine the effect of TWEAK-mediated nuclear factor (NF)-κB pathway activation in EPCs. The results revealed that TWEAK promotes EPC migration, tube formation and viability in vitro. Furthermore, TWEAK treatment resulted in improved cardiac function, decreased heart collagen and vasculogenesis in mice with AMI, which was mediated by the TWEAK- fibroblast growth factor-inducible 14 (Fn14)-NF-κB signaling pathway, as determined using Fn14 small interfering (si)RNA and Bay 11–7082 (an NF-κB inhibitor). In summary, the results of the present study suggest that activation of the TWEAK-Fn14-NF-κB signaling pathway exerts a beneficial effect on EPCs for the treatment of AMI.
Collapse
Affiliation(s)
- Zulong Sheng
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Chenwei Ju
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Bing Li
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Zhongpu Chen
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Xiaodong Pan
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Gaoliang Yan
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Yanru He
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Yuyu Yao
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
19
|
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
|
20
|
Sun XQ, Abbate A, Bogaard HJ. Role of cardiac inflammation in right ventricular failure. Cardiovasc Res 2018; 113:1441-1452. [PMID: 28957536 DOI: 10.1093/cvr/cvx159] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 08/09/2017] [Indexed: 12/18/2022] Open
Abstract
Right ventricular failure (RVF) is the main determinant of mortality in patients with pulmonary arterial hypertension (PAH). Although the exact pathophysiology underlying RVF remains unclear, inflammation may play an important role, as it does in left heart failure. Perivascular pulmonary artery and systemic inflammation is relatively well studied and known to contribute to the initiation and maintenance of the pulmonary vascular insult in PAH. However, less attention has been paid to the role of cardiac inflammation in RVF and PAH. Consistent with many other types of heart failure, cardiac inflammation, triggered by systemic and local stressors, has been shown in RVF patients as well as in RVF animal models. RV inflammation likely contributes to impaired RV contractility, maladaptive remodelling and a vicious circle between RV and pulmonary vascular injury. Although the potential to improve RV function through anti-inflammatory therapy has not been tested, this approach has been applied clinically in left ventricular failure patients, with variable success. Because inflammation plays a dual role in the development of both pulmonary vascular pathology and RVF, anti-inflammatory therapies may have a potential double benefit in patients with PAH and associated RVF.
Collapse
Affiliation(s)
- Xiao-Qing Sun
- Department of Pulmonology, VU University Medical Center/Institute for Cardiovascular Research, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Antonio Abbate
- Department of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Harm-Jan Bogaard
- Department of Pulmonology, VU University Medical Center/Institute for Cardiovascular Research, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
21
|
Zhu C, Zhang L, Liu Z, Li C, Bai Y. TWEAK/Fn14 interaction induces proliferation and migration in human airway smooth muscle cells via activating the NF-κB pathway. J Cell Biochem 2018; 119:3528-3536. [PMID: 29143982 DOI: 10.1002/jcb.26525] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/13/2017] [Indexed: 12/11/2022]
Abstract
Asthma, an increasingly common chronic disease among children, are characterized by airway remodeling, which is partly attributed to the proliferation and migration of airway smooth muscle cell (ASMC). The purpose of the present study was to investigate potential roles and mechanisms of the tumor necrosis factor-like weak inducer of apoptosis (TWEAK)/fibroblast growth factor-inducible molecule 14 (Fn14) axis on cell proliferation and migration in HASMCs. Compared to HASMCs from non-asthmatic patients, those from asthmatic patients showed elevated expression levels of both Fn14 and TWEAK. Additionally, similar to the response triggered by platelet-derived growth factor-BB, stimulation with recombinant TWEAK strongly induced cell proliferation and migration in HASMCs. However, depletion of Fn14 remarkably abrogated the enhancement of TWEAK on the cell proliferation and migration of HASMCs. Furthermore, treatment with TWEAK led to the activation of NF-κB. This effect was eliminated by silencing Fn14, indicating that TWEAK-induced NF-κB signaling was mediated via Fn14. Moreover, the TWEAK/Fn14 interaction promoted cell proliferation and migration. These effects were blocked by NF-κB inhibitor SN50, which suggest that the TWEAK/Fn14 signaling system partially depends on NF-κB activity. Collectively, we demonstrated that the TWEAK/Fn14 axis accelerated HASMC cell proliferation and migration by activating the NF-κB pathway, thereby exacerbating airway remodeling in asthma. Altogether, these findings indicate a novel role for the TWEAK/Fn14/NF-κB pathway as a potent option for limiting airway remodeling in asthma.
Collapse
Affiliation(s)
- Cuimin Zhu
- Department of Pediatrics, Cangzhou Central Hospital, Cangzhou, Hebei, PR China
| | - Leguo Zhang
- Department of Internal Neurology, Cangzhou Central Hospital, Cangzhou, Hebei, PR China
| | - Zhiming Liu
- Department of Pediatrics, Cangzhou Central Hospital, Cangzhou, Hebei, PR China
| | - Chen Li
- Department of Pediatrics, Cangzhou Central Hospital, Cangzhou, Hebei, PR China
| | - Yajie Bai
- Department of Pediatrics, Cangzhou Central Hospital, Cangzhou, Hebei, PR China
| |
Collapse
|
22
|
Involvement of TWEAK and the NF-κB signaling pathway in lupus nephritis. Exp Ther Med 2018; 15:2611-2619. [PMID: 29456665 DOI: 10.3892/etm.2018.5711] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 08/28/2017] [Indexed: 11/05/2022] Open
Abstract
Previous findings have identified that tumor necrosis factor-related weak inducer of apoptosis (TWEAK) is associated with lupus nephritis (LN) activity status; however, the mechanism involved remains unclear. The present study aimed to investigate the roles of TWEAK and the nuclear factor (NF)-κB signaling pathway in LN. TWEAK levels in the blood and urine of patients with LN or non-LN systemic lupus erythematosus were measured by ELISA and compared with those in healthy controls. TWEAK expression and NF-κB transcriptional activity in the kidney were detected by western blotting, and Ki-67 and cluster of differentiation (CD) 68 expression were assessed using immunofluorescence. Additionally, human mesangial cells (HMCs) were cultured in vitro and divided into five groups: Normal control, TWEAK stimulus group, TWEAK + TWEAK blocking antibody, TWEAK + NF-κB inhibitor (BAY 11-7082) and TWEAK + combined (blocking antibody + BAY 11-7082). Cell cycle activity and Ki-67 expression in the HMCs were evaluated using flow cytometry, and cell induction of macrophage chemotaxis was determined by a Transwell assay. Levels of the inflammation-associated factors interleukin (IL)-6, monocyte chemotactic protein 1 (MCP-1), chemokine ligand 5 (CCL5), IL-8 and IL-10 were also detected by reverse transcription-quantitative polymerase chain reaction. It was observed that the urine levels of TWEAK in patients with LN were significantly elevated compared with those in the other groups (P<0.05). LN kidneys exhibited markedly increased cell proliferative ability, macrophage infiltration, TWEAK expression and NF-κB transcriptional activity compared with normal kidneys. Furthermore, the results indicated that treatment with recombinant TWEAK notably enhanced NF-κB transcriptional activity and significantly promoted cell proliferation and cell cycle activity (P<0.05), induced macrophage chemotaxis (P<0.05), significantly increased the expression of the chemotactic factors IL-6, IL-8, MCP-1 and CCL5 (P<0.05), and significantly reduced anti-inflammatory cytokine IL-10 mRNA expression in HMCs (P<0.05), relative to normal controls. Accordingly, blocking TWEAK function or inhibiting NF-κB activity reversed these effects. Collectively these data indicate that urine TWEAK may be considered as a novel biomarker of LN activity, and that blocking TWEAK function or NF-κB activity may effectively alleviate glomerular mesangial cell proliferation and macrophage chemotaxis.
Collapse
|
23
|
Jiang L, Chen FX, Zang ST, Yang QF. Betulinic acid prevents high glucose-induced expression of extracellular matrix protein in cardiac fibroblasts by inhibiting the TGF-β1/Smad signaling pathway. Mol Med Rep 2017; 16:6320-6325. [DOI: 10.3892/mmr.2017.7323] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 03/07/2017] [Indexed: 11/06/2022] Open
|
24
|
Subramaniyan SA, Hwang I. Biological Differences between Hanwoo longissimus dorsi and semimembranosus Muscles in Collagen Synthesis of Fibroblasts. Korean J Food Sci Anim Resour 2017; 37:392-401. [PMID: 28747825 PMCID: PMC5516066 DOI: 10.5851/kosfa.2017.37.3.392] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/12/2017] [Accepted: 05/13/2017] [Indexed: 11/06/2022] Open
Abstract
Variations in physical toughness between muscles and animals are a function of growth rate and extend of collagen type I and III. The current study was designed to investigate the ability of growth rate, collagen concentration, collagen synthesizing and degrading genes on two different fibroblast cells derived from Hanwoo m. longissimus dorsi (LD) and semimembranosus (SM) muscles. Fibroblast cell survival time was determined for understanding about the characteristics of proliferation rate between the two fibroblasts. We examined the collagen concentration and protein expression of collagen type I and III between the two fibroblasts. The mRNA expression of collagen synthesis and collagen degrading genes to elucidate the molecular mechanisms on toughness and tenderness through collagen production between the two fibroblast cells. From our results the growth rate, collagen content and protein expression of collagen type I and III were significantly higher in SM than LD muscle fibroblast. The mRNA expressions of collagen synthesized genes were increased whereas the collagen degrading genes were decreased in SM than LD muscle. Results from confocal microscopical investigation showed increased fluorescence of collagen type I and III appearing stronger in SM than LD muscle fibroblast. These results implied that the locomotion muscle had higher fibroblast growth rate, leads to produce more collagen, and cause tougher than positional muscle. This in vitro study mirrored that background toughness of various muscles in live animal is likely associated with fibroblast growth pattern, collagen synthesis and its gene expression.
Collapse
Affiliation(s)
| | - Inho Hwang
- Department of Animal Science, Chonbuk National University, Jeonju 54896, Korea
| |
Collapse
|
25
|
Sequera C, Vázquez-Carballo A, Arechederra M, Fernández-Veledo S, Porras A. TWEAK promotes migration and invasion in MEFs through a mechanism dependent on ERKs activation and Fibulin 3 down-regulation. J Cell Physiol 2017; 233:968-978. [PMID: 28383766 DOI: 10.1002/jcp.25942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 03/30/2017] [Indexed: 11/10/2022]
Abstract
TWEAK regulates multiple physio-pathological processes in fibroblasts such as fibrosis. It also induces migration and invasion in tumors and it can activate p38 MAPK in various cell types. Moreover, p38α MAPK promotes migration and invasion in several cancer cells types and in mouse embryonic fibroblasts (MEFs). However, it remains unknown if TWEAK could promote migration in fibroblasts and whether p38α MAPK might play a role. Our results reveal that TWEAK activates ERKs, Akt, and p38α/β MAPKs and reduces secreted Fibulin 3 in MEFs. TWEAK also increases migration and invasion in wt and p38α deficient MEFs, which indicates that p38α MAPK is not required to mediate these effects. In contrast, ERKs inhibition significantly decreases TWEAK-induced migration and Fibulin 3 knock-down mimics TWEAK effect. These results indicate that both ERKs activation and Fibulin 3 down-regulation would contribute to mediate TWEAK pro-migratory effect. In fact, the additional regulation of ERKs and/or p38β as a consequence of Fibulin 3 decrease might be also involved in the pro-migratory effect of TWEAK in MEFs. In conclusion, our studies uncover novel mechanisms by which TWEAK would favor tissue repair by promoting fibroblasts migration.
Collapse
Affiliation(s)
- Celia Sequera
- Facultad de Farmacia, Departamento de Bioquímica y Biología Molecular II, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Ana Vázquez-Carballo
- Facultad de Farmacia, Departamento de Bioquímica y Biología Molecular II, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - María Arechederra
- Facultad de Farmacia, Departamento de Bioquímica y Biología Molecular II, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Sonia Fernández-Veledo
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain.,Hospital Universitari de Tarragona Joan XXIII, IISPV, Universitat Rovira i Virgili, Tarragona, Spain
| | - Almudena Porras
- Facultad de Farmacia, Departamento de Bioquímica y Biología Molecular II, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| |
Collapse
|
26
|
Electrical Stimulation Based on Chronaxie Increases Fibrosis and Modulates TWEAK/Fn14, TGF-β/Myostatin, and MMP Pathways in Denervated Muscles. Am J Phys Med Rehabil 2017; 96:260-267. [DOI: 10.1097/phm.0000000000000601] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
27
|
Martínez-Miguel P, Medrano-Andrés D, Griera-Merino M, Ortiz A, Rodríguez-Puyol M, Rodríguez-Puyol D, López-Ongil S. Tweak up-regulates endothelin-1 system in mouse and human endothelial cells. Cardiovasc Res 2016; 113:207-221. [DOI: 10.1093/cvr/cvw239] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 10/20/2016] [Accepted: 11/21/2016] [Indexed: 12/21/2022] Open
|
28
|
Basso D, Padoan A, Laufer T, Aneloni V, Moz S, Schroers H, Pelloso M, Saiz A, Krapp M, Fogar P, Cornoldi P, Zambon CF, Rossi E, La Malfa M, Marotti A, Brefort T, Weis TM, Katus HA, Plebani M. Relevance of pre-analytical blood management on the emerging cardiovascular protein biomarkers TWEAK and HMGB1 and on miRNA serum and plasma profiling. Clin Biochem 2016; 50:186-193. [PMID: 27847340 DOI: 10.1016/j.clinbiochem.2016.11.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 11/07/2016] [Accepted: 11/08/2016] [Indexed: 01/07/2023]
Abstract
BACKGROUND Disease-independent sources of biomarker variability include pre-analytical, analytical and biological variance. The aim of the present study was to evaluate whether the pre-analytical phase has any impact on the emerging heart disease TWEAK and HMGB1 protein markers and miRNA biomarkers, and whether peptidome profiling allows the identification of pre-analytical quality markers. METHODS An assessment was made of sample type (serum, EDTA-Plasma, Citrate-Plasma, ACD-plasma, Heparin-plasma), temperature of sample storage (room temperature or refrigerated), time of sample storage (0.5, 3, 6 and 9h) and centrifugation (one or two-step). Aliquots of all processed samples were immediately frozen (-80°C) before analysis. Proteins were assayed by ELISAs, miRNA expression profile by microarray and peptidome profiling by MALDI-TOF/MS. RESULTS Temperature, time and centrifugation had no impact on TWEAK and HMGB1 results, which were significantly influenced by matrix type, TWEAK levels being significantly higher (F=194.7, p<0.0001), and HMGB1 levels significantly lower (F=36.32, p<0.0001) in serum than in any other plasma type. Unsuitable miRNA results were obtained using Heparin-plasma. Serum miRNA expression profiles depended mainly on temperature, while EDTA-plasma miRNA expression profiles were strongly affected by the centrifugation method used. MALDI-TOF/MS allowed the identification of seven features as indices of pre-analytical serum (m/z at 1206, 1350, 1865 and 2021) or EDTA-plasma (m/z 1897, 2740 and 2917) degradation. CONCLUSIONS Serum and EDTA-plasma allow the analysis of both proteins and miRNA emerging biomarkers of heart diseases. Refrigerated storage prevents an altered miRNA expression profile also in cases of a prolonged time-interval between blood drawing and processing.
Collapse
Affiliation(s)
- Daniela Basso
- Department of Medicine - DIMED, University of Padova, Italy.
| | - Andrea Padoan
- Department of Medicine - DIMED, University of Padova, Italy
| | - Thomas Laufer
- Comprehensive Biomarker Center GmbH, Heidelberg, Germany
| | | | - Stefania Moz
- Department of Medicine - DIMED, University of Padova, Italy
| | | | | | - Anna Saiz
- Comprehensive Biomarker Center GmbH, Heidelberg, Germany
| | - Medea Krapp
- Comprehensive Biomarker Center GmbH, Heidelberg, Germany
| | - Paola Fogar
- Department of Medicine - DIMED, University of Padova, Italy
| | - Paola Cornoldi
- Department of Medicine - DIMED, University of Padova, Italy
| | | | - Elisa Rossi
- Department of Medicine - DIMED, University of Padova, Italy
| | - Marco La Malfa
- Department of Medicine - DIMED, University of Padova, Italy
| | - Alberto Marotti
- UOC Immunotrasfusionale, University-Hospital of Padova, Italy
| | - Thomas Brefort
- Comprehensive Biomarker Center GmbH, Heidelberg, Germany; Eurofins Medigenomix GmbH, Ebersberg, Germany
| | - Tanja M Weis
- Department of Cardiology, Angiology, and Pulmonology, University Hospital Heidelberg, Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, Angiology, and Pulmonology, University Hospital Heidelberg, Heidelberg, Germany
| | - Mario Plebani
- Department of Medicine - DIMED, University of Padova, Italy
| |
Collapse
|
29
|
Xu J, He J, He H, Peng R, Xi J. TWEAK-Fn14 Influences Neurogenesis Status via Modulating NF-κB in Mice with Spinal Cord Injury. Mol Neurobiol 2016; 54:7497-7506. [PMID: 27822714 DOI: 10.1007/s12035-016-0248-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 10/23/2016] [Indexed: 01/01/2023]
Abstract
The aim of our research is to investigate the regulatory role of TNF-like weak inducer of apoptosis- fibroblast growth factor-inducible 14 (TWEAK-Fn14) pathway in nuclear factor-kappa B (NF-κB) expression and neurogenesis status after spinal cord injury (SCI). We constructed a mice model of spinal cord injury and injected different lentiviral vectors which were transfected with TWEAK, TWEAK small interfering RNA (siRNA) and Fn14 siRNA into different groups of mice. Locomotor functional recovery status of the hind limb in mice was assessed using the Basso, Beattie and Bresnahan (BBB) test. Apoptosis status in the injured area was examined via TDT-mediated dUTP-biotin nick end-labeling (TUNEL) staining, the expression of GAP-43 in injured spinal cord was quantified by immunohistochemistry and expressions of TWEAK, Fn14, NF-κB, TNF-α, and IL-1β were evaluated by either western blot or ELISA. The expressions of TWEAK, Fn14, and NF-κB in the model group were significantly higher compared with those in the control group. Furthermore, the TWEAK group in which TWEAK was overexpressed exhibited significantly higher expressions of TWEAK, Fn14, and NF-κB, TNF-α and IL-1β in relation to those in the model group (P < 0.05 for all). Moreover, the transfection of Fn14 siRNA antagonized the above effect of TWEAK transfection on injured mice. On the other hand, the TWEAK siRNA group in which the expression of TWEAK was inhibited exhibited significantly lower expressions of TWEAK, Fn14, NF-κB, TNF-α, and IL-1β (P < 0.05 for all). Moreover, the transfection of TWEAK siRNA enhanced the locomotor functional recovery status in injured mice and suppressed the apoptosis of injured areas (P < 0.05 for all). In conclusion, stimulating the TWEAK-Fn14 pathway may elevate the expression of NF-κB, thereby slow the function recovery of SCI mice whereas inhibiting the TWEAK-Fn14 pathway may improve the neurogenesis status in mice with spinal cord injuries.
Collapse
Affiliation(s)
- Jing Xu
- Department of Otolaryngology-Head and Neck Surgery, Xiang-Ya Hospital, Central South University, Changsha, Hunan, 410008, China.,Otolaryngology Key Laboratory of Hunan Province, Changsha, Hunan, 410008, China
| | - Jian He
- Department of Otolaryngology-Head and Neck Surgery, Xiang-Ya Hospital, Central South University, Changsha, Hunan, 410008, China.,Otolaryngology Key Laboratory of Hunan Province, Changsha, Hunan, 410008, China
| | - Huang He
- Department of Neurosurgery, Xiang-Ya Hospital, Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, China
| | - Renjun Peng
- Department of Neurosurgery, Xiang-Ya Hospital, Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, China
| | - Jian Xi
- Department of Neurosurgery, Xiang-Ya Hospital, Central South University, No. 87 Xiangya Road, Changsha, Hunan, 410008, China.
| |
Collapse
|
30
|
Abstract
The mechanisms underlying hepatic inflammation and fibrogenesis in chronic hepatitis B (CHB) are complex and several cytokines are involved. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is a member of the tumor necrosis factor superfamily which also acts as a cytokine. This study was conducted to evaluate serum soluble TWEAK (sTWEAK) levels in noncirrhotic CHB patients.Fifty-two treatment naive CHB patients and 30 healthy controls were included in the study and serum sTWEAK concentrations were measured using commercially available ELISA kits.Mean serum sTWEAK concentration was significantly lower in CHB group than healthy controls (189.6 ± 63.3 pg/mL in CHB group and 297.6 ± 61.5 pg/mL in control group, P < 0.001). According to the degree of necroinflammation in liver biopsies mean sTWEAK concentrations were found to be 168.14 ± 51.51, 206.96 ± 58.51, and 223.62 ± 78.88 pg/mL in patients with mild, moderate, and severe inflammation, respectively, and the difference between groups was statistically significant (P = 0.022). sTWEAK concentration was also found to be significantly higher in patients with advanced fibrosis in liver biopsy samples (169.59 ± 52.02 and 211.17 ± 68.22 pg/mL in patients with mild and advanced fibrosis, respectively, P = 0.016). Receiver operating characteristic (ROC) curves were obtained in CHB group to differentiate patients with advanced fibrosis from patients with mild fibrosis. Area under curve (AUC) was 0.676 (95% Cl; 0.526-0.825) for sTWEAK and for the specified cut-off value of 213.67 pg/mL sensitivity and specificity were 60% and 81.4%, respectively. ROC curve for sTWEAK to differentiate patients with severe inflammation revealed an AUC of 0.664 (95% Cl; 0.450-0.878). A cut-off value of 243.27 pg/mL yielded 54.5% sensitivity and 82.9% specificity.Serum sTWEAK concentration is decreased in treatment naive CHB patients. Further studies with simultaneous determination of circulating sTWEAK concentrations and TWEAK and factor-inducible 14 (Fn14) expressions in the liver biopsy samples would clarify the exact association of TWEAK/Fn14 pathway in the pathogenesis of CHB.
Collapse
Affiliation(s)
- Mehmet Asil
- Division of Gastroenterology, Department of Internal Medicine, Meram School of Medicine, Necmettin Erbakan University, Meram, Konya, Turkey
- Correspondence: Mehmet Asil, Necmettin Erbakan Üniversitesi, Meram Tıp Fakültesi, İç Hastalıkları Anabilim, Dalı, Gastroenteroloji Kliniği, 42090 Meram, Konya, Turkey (e-mail: )
| | | |
Collapse
|
31
|
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
|
32
|
Shang H, Wang Z, Song Y. Liver progenitor cells-mediated liver regeneration in liver cirrhosis. Hepatol Int 2016; 10:440-447. [PMID: 26742763 DOI: 10.1007/s12072-015-9693-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 12/07/2015] [Indexed: 02/08/2023]
Abstract
Cirrhosis is defined as the histological development of regenerative nodules surrounded by fibrous bands in response to chronic liver injury. In cirrhotic liver where hepatocytes proliferation is compromised, liver progenitor cells (LPCs) are activated and then differentiated into hepatocytes and cholangiocytes, leading to the generation of regenerative nodules and functional restoration. Here, we summarize and discuss recent findings on the mechanisms underlying LPCs-mediated regeneration in liver cirrhosis. Firstly, we provide recent research on the mechanism underlying LPCs activation in severe or chronic liver injury. Secondly, we present new and exciting data on exploring the origin of LPCs, which reveal that the hepatocytes give rise to duct-like progenitors that then differentiate back into hepatocytes in chronic liver injury or liver cirrhosis. Finally, we highlight recent findings from the literature exploring the role of LPCs niche in directing the behavior and fate of LPCs. This remarkable insight into the cellular and molecular mechanisms of LPCs-mediated regeneration in liver cirrhosis will provide a basis for translating this knowledge into clinical application.
Collapse
Affiliation(s)
- Haitao Shang
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, 256603, People's Republic of China
| | - Zhijun Wang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Yuhu Song
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
| |
Collapse
|
33
|
Wilhelm A, Shepherd EL, Amatucci A, Munir M, Reynolds G, Humphreys E, Resheq Y, Adams DH, Hübscher S, Burkly LC, Weston CJ, Afford SC. Interaction of TWEAK with Fn14 leads to the progression of fibrotic liver disease by directly modulating hepatic stellate cell proliferation. J Pathol 2016; 239:109-21. [PMID: 26924336 PMCID: PMC4949530 DOI: 10.1002/path.4707] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 01/31/2016] [Accepted: 02/17/2016] [Indexed: 12/21/2022]
Abstract
Tumour necrosis factor‐like weak inducer of apoptosis (TWEAK) and its receptor fibroblast growth factor‐inducible 14 (Fn14) have been associated with liver regeneration in vivo. To further investigate the role of this pathway we examined their expression in human fibrotic liver disease and the effect of pathway deficiency in a murine model of liver fibrosis. The expression of Fn14 and TWEAK in normal and diseased human and mouse liver tissue and primary human hepatic stellate cells (HSCs) were investigated by qPCR, western blotting and immunohistochemistry. In addition, the levels of Fn14 in HSCs following pro‐fibrogenic and pro‐inflammatory stimuli were assessed and the effects of exogenous TWEAK on HSCs proliferation and activation were studied in vitro. Carbon tetrachloride (CCl4) was used to induce acute and chronic liver injury in TWEAK KO mice. Elevated expression of both Fn14 and TWEAK were detected in acute and chronic human liver injury, and co‐localized with markers of activated HSCs. Fn14 levels were low in quiescent HSCs but were significantly induced in activated HSCs, which could be further enhanced with the profibrogenic cytokine TGFβin vitro. Stimulation with recombinant TWEAK induced proliferation but not further HSCs activation. Fn14 gene expression was also significantly up‐regulated in CCl4 models of hepatic injury whereas TWEAK KO mice showed reduced levels of liver fibrosis following chronic CCl4 injury. In conclusion, TWEAK/Fn14 interaction leads to the progression of fibrotic liver disease via direct modulation of HSCs proliferation, making it a potential therapeutic target for liver fibrosis. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Annika Wilhelm
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK
| | - Emma L Shepherd
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK
| | - Aldo Amatucci
- Department of Immunology, Biogen, Cambridge, MA, USA
| | - Mamoona Munir
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK
| | - Gary Reynolds
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK
| | - Elizabeth Humphreys
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK
| | - Yazid Resheq
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK.,Medizinische Klinik 5/Department of Internal Medicine 5, Universitätsklinikum Erlangen/University Medical Centre Erlangen, Germany
| | - David H Adams
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK
| | - Stefan Hübscher
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK.,Department of Cellular Pathology, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK
| | | | - Christopher J Weston
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK
| | - Simon C Afford
- Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, UK
| |
Collapse
|
34
|
Chen S, Liu J, Yang M, Lai W, Ye L, Chen J, Hou X, Ding H, Zhang W, Wu Y, Liu X, Huang S, Yu X, Xiao D. Fn14, a Downstream Target of the TGF-β Signaling Pathway, Regulates Fibroblast Activation. PLoS One 2015; 10:e0143802. [PMID: 26625141 PMCID: PMC4666639 DOI: 10.1371/journal.pone.0143802] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 11/10/2015] [Indexed: 12/25/2022] Open
Abstract
Fibrosis, the hallmark of human injuries and diseases such as serious burns, is characterized by excessive collagen synthesis and myofibroblast accumulation. Transforming growth factor-β (TGF-β), a potent inducer of collagen synthesis, has been implicated in fibrosis in animals. In addition to TGF-β, fibroblast growth factor-inducible molecule 14 (Fn14) has been reported to play an important role in fibrotic diseases, such as cardiac fibrosis. However, the function and detailed regulatory mechanism of Fn14 in fibrosis are unclear. Here, we investigated the effect of Fn14 on the activation of human dermal fibroblasts. In normal dermal fibroblasts, TGF-β signaling increased collagen production and Fn14 expression. Furthermore, Fn14 siRNA blocked extracellular matrix gene expression; even when TGF-β signaling was activated by TGF-β1, fibroblast activation remained blocked in the presence of Fn14 siRNA. Overexpressing Fn14 increased extracellular matrix gene expression. In determining the molecular regulatory mechanism, we discovered that SMAD4, an important TGF-β signaling co-mediator, bound to the Fn14 promoter and activated Fn14 transcription. Taken together, these results indicate that the TGF-β signaling pathway activates Fn14 expression through the transcription factor SMAD4 and that activated Fn14 expression increases extracellular matrix synthesis and fibroblast activation. Therefore, Fn14 may represent a promising approach to preventing the excessive accumulation of collagen or ECM in skin fibrosis.
Collapse
Affiliation(s)
- Shaoxian Chen
- Medical Research Department of Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Juli Liu
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Min Yang
- Medical Research Department of Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- Pharmacy Department of Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Wen Lai
- Burn and Wound Repair Surgery of Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Litong Ye
- Pharmacy Department of General Hospital of Guangzhou Military Command of PLA, Guangzhou, Guangdong, China
| | - Jing Chen
- Medical Research Department of Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Xinghua Hou
- Medical Research Department of Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Hong Ding
- Medical Research Department of Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Wenwei Zhang
- Medical Research Department of Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yueheng Wu
- Medical Research Department of Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Xiaoying Liu
- Medical Research Department of Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Shufang Huang
- Medical Research Department of Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Xiyong Yu
- Medical Research Department of Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- * E-mail: (DX); (XY)
| | - Dingzhang Xiao
- Medical Research Department of Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- * E-mail: (DX); (XY)
| |
Collapse
|
35
|
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
|
36
|
Li W, Xu Q, Deng Y, Yang Z, Xing S, Zhao X, Zhu P, Wang X, He Z, Gao Y. High-mobility group box 1 accelerates lipopolysaccharide-induced lung fibroblast proliferation in vitro: involvement of the NF-κB signaling pathway. J Transl Med 2015; 95:635-47. [PMID: 25867768 DOI: 10.1038/labinvest.2015.44] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 01/18/2023] Open
Abstract
The mechanism underlying lipopolysaccharide (LPS)-induced aberrant proliferation of lung fibroblasts in Gram-negative bacilli-associated pulmonary fibrosis is unknown. High-mobility group box 1 (HMGB1) is a ubiquitous nuclear protein that is released from the nuclei of lung fibroblasts after LPS stimulation. It can exasperate LPS-induced inflammation and hasten cell proliferation. Thus, this study investigated the effects of LPS- and/or HMGB1-stimulating murine lung fibroblasts on gene expression using various assays in vitro. Thiazolyl-diphenyl-tetrazolium bromide (MTT) assay data showed that either LPS or HMGB1 could induce lung fibroblast proliferation. Endogenous HMGB1 secreted from lung fibroblasts was detected by enzyme-linked immunosorbent assay (ELISA) 48 h after LPS stimulation. Pretreatment with an anti-HMGB1 antibody inhibited the proliferative effects of LPS on lung fibroblasts. DNA microarray data showed that the NF-κB signaling genes were upregulated in cells after stimulated with LPS, HMGB1, or both. Secretion of matrix metalloproteinase (MMP)-2 and MMP-9, and tissue inhibitor of metalloproteinase 2 (TIMP-2) was significantly upregulated after treatment with LPS, HMGB1, or their combination. However, an NF-κB inhibitor was able to downregulate levels of these proteins. In addition, levels of Toll-like receptor 4 (TLR4), Toll-like receptor 2 (TLR2), and receptors for advanced glycation end products (RAGE) mRNA and proteins were also upregulated in these cells after LPS treatment and further upregulated by LPS plus HMGB1. In conclusion, the data from the current study demonstrate that LPS-induced lung fibroblast secretion of endogenous HMGB1 can augment the proproliferative effects of LPS and, therefore, may play a key role in exacerbation of pulmonary fibrosis. The underlying molecular mechanisms are related to the activation of the TLR4/NF-κB signaling pathway and its downstream targets.
Collapse
Affiliation(s)
- Wen Li
- Department of Critical Care Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiaoyi Xu
- Department of Critical Care Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuxiao Deng
- Department of Critical Care Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhongwei Yang
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shunpeng Xing
- Department of Critical Care Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xianyuan Zhao
- Department of Critical Care Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Zhu
- Department of Critical Care Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiangrui Wang
- Department of Anesthesiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhengyu He
- Department of Critical Care Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan Gao
- Department of Critical Care Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
37
|
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
|
38
|
Song J, Zhu Y, Li J, Liu J, Gao Y, Ha T, Que L, Liu L, Zhu G, Chen Q, Xu Y, Li C, Li Y. Pellino1-mediated TGF-β1 synthesis contributes to mechanical stress induced cardiac fibroblast activation. J Mol Cell Cardiol 2014; 79:145-56. [PMID: 25446187 DOI: 10.1016/j.yjmcc.2014.11.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 10/22/2014] [Accepted: 11/04/2014] [Indexed: 11/24/2022]
Abstract
Activation of cardiac fibroblasts is a key event in the progression of cardiac fibrosis that leads to heart failure. However, the molecular mechanisms underlying mechanical stress-induced cardiac fibroblast activation are complex and poorly understood. This study demonstrates that Pellino1, an E3 ubiquitin ligase, was activated in vivo in pressure overloaded rat hearts and in cultured neonatal rat cardiac fibroblasts (NRCFs) exposed to mechanical stretch in vitro. Suppression of the expression and activity of Pellino1 by adenovirus-mediated delivery of shPellino1 (adv-shpeli1) attenuated pressure overload-induced cardiac dysfunction and cardiac hypertrophy and decreased cardiac fibrosis in rat hearts. Transfection of adv-shpeli1 also significantly attenuated mechanical stress-induced proliferation, differentiation and collagen synthesis in NRCFs. Pellino1 silencing also abrogated mechanical stretch-induced polyubiquitination of tumor necrosis factor-alpha receptor association factor-6 (TRAF6) and receptor-interacting protein 1 (RIP1) and consequently decreased the DNA binding activity of nuclear factor-kappa B (NF-κB) in NRCFs. In addition, Pellino1 silencing prevented stretch-induced activation of p38 and activator protein 1 (AP-1) binding activity in NRCFs. Chromatin Immunoprecipitation (ChIP) and luciferase reporter assays showed that Pellino1 silencing prevented the binding of NF-κB and AP-1 to the promoter region of transforming growth factor-β1 (TGF-β1) thus dampening TGF-β1 transactivation. Our data reveal a previously unrecognized role of Pellino1 in extracellular matrix deposition and cardiac fibroblast activation in response to mechanical stress and provides a novel target for treatment of cardiac fibrosis and heart failure.
Collapse
Affiliation(s)
- Juan Song
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, Jiangsu, China
| | - Yun Zhu
- Department of Pathology, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, Jiangsu, China
| | - Jiantao Li
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, Jiangsu, China
| | - Jiahao Liu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, Jiangsu, China
| | - Yun Gao
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, Jiangsu, China
| | - Tuanzhu Ha
- Department of Surgery, East Tennessee State University, Campus Box 70575, Johnson City, TN 37614-0575, USA
| | - Linli Que
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, Jiangsu, China
| | - Li Liu
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Guoqing Zhu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Qi Chen
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, Jiangsu, China
| | - Yong Xu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, Jiangsu, China
| | - Chuanfu Li
- Department of Surgery, East Tennessee State University, Campus Box 70575, Johnson City, TN 37614-0575, USA
| | - Yuehua Li
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology, Nanjing Medical University, 140 Hanzhong Road, Nanjing 210029, Jiangsu, China.
| |
Collapse
|
39
|
Katoonizadeh A, Poustchi H, Malekzadeh R. Hepatic progenitor cells in liver regeneration: current advances and clinical perspectives. Liver Int 2014; 34:1464-1472. [PMID: 24750779 DOI: 10.1111/liv.12573] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 04/17/2014] [Indexed: 12/12/2022]
Abstract
When there is a massive loss of hepatocytes and/or an inhibition in the proliferative capacity of the mature hepatocytes, activation of a dormant cell population of resident hepatic progenitor cells (HPCs) occurs. Depending on the type of liver damage HPCs generate new hepatocytes and biliary cells to repopulate the liver placing them as potential candidates for cell therapy in human liver failure. Liver injury specific mechanisms through which HPCs differentiate towards mature epithelial cell types are recently become understood. Such new insights will enable us not only to direct HPCs behaviour for therapeutic purposes, but also to develop clinically feasible methods for in vivo differentiation of other stem cell types towards functional hepatocytes. This review aimed to provide the current improved knowledge of the role of HPCs niche and its signals in directing the behaviour and fate of HPCs and to translate this basic knowledge of HPCs activation/differentiation into its clinical applications.
Collapse
Affiliation(s)
- Aezam Katoonizadeh
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | | |
Collapse
|
40
|
Wajant H. The TWEAK-Fn14 system as a potential drug target. Br J Pharmacol 2014; 170:748-64. [PMID: 23957828 DOI: 10.1111/bph.12337] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 07/29/2013] [Accepted: 08/12/2013] [Indexed: 12/27/2022] Open
Abstract
Fibroblast growth factor-inducible 14 (Fn14) is a member of the tumour necrosis factor (TNF) receptor family that is induced in a variety of cell types in situations of tissue injury. Fn14 becomes activated by TNF-like weak inducer of apoptosis (TWEAK), a typical member of the TNF ligand family. TWEAK is constitutively expressed by monocytes and some tumour cell lines and also shows cytokine inducible expression in various other cell types. Fn14 activation results in stimulation of signalling pathways culminating in the activation of NFκB transcription factors and various MAPKs but might also trigger the PI3K/Akt pathway and GTPases of the Rho family. In accordance with its tissue damage-associated expression pattern and its pleiotropic proinflammatory signalling capabilities, the TWEAK-Fn14 system has been implicated in a huge number of pathologies. The use of TWEAK- and Fn14-knockout mice identified the TWEAK-Fn14 system as a crucial player in muscle atrophy, cerebral ischaemia, kidney injury, atherosclerosis and infarction as well as in various autoimmune scenarios including experimental autoimmune encephalitis, rheumatoid arthritis and inflammatory bowel disease. Moreover, there is increasing preclinical evidence that Fn14 targeting is a useful option in tumour therapy. Based on a discussion of the signalling capabilities of TWEAK and Fn14, this review is focused on two major issues. On the one hand, on the molecular and cellular basis of the TWEAK/Fn14-related pathological outcomes in the aforementioned diseases and on the other hand, on the preclinical experience that have been made so far with TWEAK and Fn14 targeting drugs.
Collapse
Affiliation(s)
- Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW The tumor necrosis factor-like weak inducer of apoptosis (TWEAK) cytokine has been linked to kidney injury by functional studies in experimental animals, and has biomarker potential in kidney disease. RECENT FINDINGS TWEAK was known to promote tubular cell injury and kidney inflammation. Recent studies have expanded these observations, identifying additional targets of TWEAK relevant to kidney injury. Thus, TWEAK upregulates the chemokine and cholesterol scavenger receptor CXCL16 and downregulates the antiaging and antifibrotic molecule Klotho in tubular cells. Furthermore, fibrogenic TWEAK actions on renal fibroblasts were described. TWEAK or factor-inducible molecule 14 targeting decreased the kidney fibrosis resulting from immune and nonimmune kidney injury induced by transient tubular or glomerular insults or by persistent urinary tract obstruction. TWEAK might also contribute to the link between chronic kidney disease and kidney cancer, as suggested by its role in other genitourinary cancers. Progress has also been made in TWEAK targeting. A phase I clinical trial showed that TWEAK targeting is well tolerated in humans, and an ongoing trial is exploring efficacy in lupus nephritis. Nanomolecules and inhibitors of epidermal growth factor receptor pathway may also protect from the adverse effects of TWEAK in the kidney. SUMMARY These findings suggest that TWEAK targeting has clinical potential in kidney injury of immune and nonimmune origin.
Collapse
|
42
|
Martín P, Mora I, Cortes MA, Calleros L, García-Jerez A, Ortiz A, Rodríguez-Puyol M, Rodríguez-Puyol D, Olmos G. Relevant role of PKG in the progression of fibrosis induced by TNF-like weak inducer of apoptosis. Am J Physiol Renal Physiol 2014; 307:F75-85. [DOI: 10.1152/ajprenal.00398.2013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
TNF-like weak inducer of apoptosis (TWEAK) is an inflammatory cytokine that activates the FGF-inducible 14 receptor. Both TWEAK and the FGF-inducible 14 receptor are constitutively expressed in the kidney. TWEAK has been shown to modulate several biological responses, such as inflammation, proliferation, differentiation, and apoptosis, that contribute to kidney injury. However, the role of TWEAK in fibrosis and TWEAK-activated intracellular signaling pathways remain poorly understood. We tested the hypothesis that TWEAK can be a potent inducer of renal fibrosis by increasing transforming growth factor (TGF)-β1 expression (a well-known switch in the fibrosis process) through PKG-I downregulation. We showed that in human mesangial cells, TWEAK increased TGF-β1 expression and activity, leading to higher levels of the extracellular matrix protein fibronectin and decreased PKG-I expression and activity via the Ras pathway. PKG-I activation with 8-bromo-cGMP, Ras inactivation with dominant negative Ras, or Ras pathway inhibition with the ERK1/2 inhibitor PD-98059 resulted in the prevention of TWEAK-induced TGF-β1 upregulation. In vivo, exogenous administration of TWEAK to wild-type mice downregulated kidney PKG-I and increased kidney TGF-β1 expression. These effects were blunted in H-Ras knockout mice. Together, these data demonstrate, for the first time, the key role of PKG-I in TGF-β1 induction by TWEAK in kidney cells.
Collapse
Affiliation(s)
- Paloma Martín
- Department of System Biology, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
- REDinREN (Instituto de Salud Carlos III), Madrid, Spain
- Instituto Reina Sofía de Investigaciones Nefrológicas, Madrid, Spain
| | - Inés Mora
- REDinREN (Instituto de Salud Carlos III), Madrid, Spain
- Instituto Reina Sofía de Investigaciones Nefrológicas, Madrid, Spain
| | - M. Alicia Cortes
- Department of System Biology, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
- REDinREN (Instituto de Salud Carlos III), Madrid, Spain
- Instituto Reina Sofía de Investigaciones Nefrológicas, Madrid, Spain
| | - Laura Calleros
- Department of System Biology, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
- REDinREN (Instituto de Salud Carlos III), Madrid, Spain
- Instituto Reina Sofía de Investigaciones Nefrológicas, Madrid, Spain
| | - Andrea García-Jerez
- Department of System Biology, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
- REDinREN (Instituto de Salud Carlos III), Madrid, Spain
- Instituto Reina Sofía de Investigaciones Nefrológicas, Madrid, Spain
| | - Alberto Ortiz
- REDinREN (Instituto de Salud Carlos III), Madrid, Spain
- IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Rodríguez-Puyol
- Department of System Biology, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
- REDinREN (Instituto de Salud Carlos III), Madrid, Spain
- Instituto Reina Sofía de Investigaciones Nefrológicas, Madrid, Spain
| | - Diego Rodríguez-Puyol
- REDinREN (Instituto de Salud Carlos III), Madrid, Spain
- Instituto Reina Sofía de Investigaciones Nefrológicas, Madrid, Spain
- Department of Medicine, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
- Nephrology Section and Research Unit, Hospital Príncipe de Asturias, Alcalá de Henares, Madrid, Spain; and
| | - Gemma Olmos
- Department of System Biology, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
- REDinREN (Instituto de Salud Carlos III), Madrid, Spain
- Instituto Reina Sofía de Investigaciones Nefrológicas, Madrid, Spain
| |
Collapse
|
43
|
Novoyatleva T, Sajjad A, Engel FB. TWEAK-Fn14 Cytokine-Receptor Axis: A New Player of Myocardial Remodeling and Cardiac Failure. Front Immunol 2014; 5:50. [PMID: 24611063 PMCID: PMC3920183 DOI: 10.3389/fimmu.2014.00050] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 01/28/2014] [Indexed: 01/01/2023] Open
Abstract
Tumor necrosis factor (TNF) has been firmly established as a pathogenic factor in heart failure, a significant socio-economic burden. In this review, we will explore the role of other members of the TNF/TNF receptor superfamily (TNFSF/TNFRSF) in cardiovascular diseases (CVDs) focusing on TWEAK and its receptor Fn14, new players in myocardial remodeling and heart failure. The TWEAK/Fn14 pathway controls a variety of cellular activities such as proliferation, differentiation, and apoptosis and has diverse biological functions in pathological mechanisms like inflammation and fibrosis that are associated with CVDs. Furthermore, it has recently been shown that the TWEAK/Fn14 axis is a positive regulator of cardiac hypertrophy and that deletion of Fn14 receptor protects from right heart fibrosis and dysfunction. We discuss the potential use of the TWEAK/Fn14 axis as biomarker for CVDs as well as therapeutic target for future treatment of human heart failure based on supporting data from animal models and in vitro studies. Collectively, existing data strongly suggest the TWEAK/Fn14 axis as a potential new therapeutic target for achieving cardiac protection in patients with CVDs.
Collapse
Affiliation(s)
- Tatyana Novoyatleva
- Department of Cardiac Development and Remodelling, Max-Planck-Institute for Heart and Lung Research , Bad Nauheim , Germany
| | - Amna Sajjad
- Department of Cardiac Development and Remodelling, Max-Planck-Institute for Heart and Lung Research , Bad Nauheim , Germany ; Government College University Faisalabad , Faisalabad , Pakistan
| | - Felix B Engel
- Department of Nephropathology, Experimental Renal and Cardiovascular Research, Institute of Pathology, University of Erlangen-Nürnberg , Erlangen , Germany
| |
Collapse
|
44
|
Enwere EK, Lacasse EC, Adam NJ, Korneluk RG. Role of the TWEAK-Fn14-cIAP1-NF-κB Signaling Axis in the Regulation of Myogenesis and Muscle Homeostasis. Front Immunol 2014; 5:34. [PMID: 24550918 PMCID: PMC3913901 DOI: 10.3389/fimmu.2014.00034] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 01/21/2014] [Indexed: 12/16/2022] Open
Abstract
Mammalian skeletal muscle maintains a robust regenerative capacity throughout life, largely due to the presence of a stem cell population known as “satellite cells” in the muscle milieu. In normal conditions, these cells remain quiescent; they are activated upon injury to become myoblasts, which proliferate extensively and eventually differentiate and fuse to form new multinucleated muscle fibers. Recent findings have identified some of the factors, including the cytokine TNFα-like weak inducer of apoptosis (TWEAK), which govern these cells’ decisions to proliferate, differentiate, or fuse. In this review, we will address the functions of TWEAK, its receptor Fn14, and the associated signal transduction molecule, the cellular inhibitor of apoptosis 1 (cIAP1), in the regulation of myogenesis. TWEAK signaling can activate the canonical NF-κB signaling pathway, which promotes myoblast proliferation and inhibits myogenesis. In addition, TWEAK activates the non-canonical NF-κB pathway, which, in contrast, promotes myogenesis by increasing myoblast fusion. Both pathways are regulated by cIAP1, which is an essential component of downstream signaling mediated by TWEAK and similar cytokines. This review will focus on the seemingly contradictory roles played by TWEAK during muscle regeneration, by highlighting the interplay between the two NF-κB pathways under physiological and pathological conditions. We will also discuss how myogenesis is negatively affected by chronic conditions, which affect homeostasis of the skeletal muscle environment.
Collapse
Affiliation(s)
- Emeka K Enwere
- Department of Medical Microbiology and Immunology, University of Alberta , Edmonton, AB , Canada
| | - Eric C Lacasse
- Solange Gauthier Karsh Molecular Genetics Laboratory, Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute , Ottawa, ON , Canada
| | - Nadine J Adam
- Solange Gauthier Karsh Molecular Genetics Laboratory, Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute , Ottawa, ON , Canada ; Department of Biochemistry, Microbiology and Immunology, University of Ottawa , Ottawa, ON , Canada
| | - Robert G Korneluk
- Solange Gauthier Karsh Molecular Genetics Laboratory, Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute , Ottawa, ON , Canada ; Department of Biochemistry, Microbiology and Immunology, University of Ottawa , Ottawa, ON , Canada
| |
Collapse
|
45
|
Sastre C, Fernández-Laso V, Madrigal-Matute J, Muñoz-García B, Moreno JA, Pastor-Vargas C, Llamas-Granda P, Burkly LC, Egido J, Martín-Ventura JL, Blanco-Colio LM. Genetic deletion or TWEAK blocking antibody administration reduce atherosclerosis and enhance plaque stability in mice. J Cell Mol Med 2014; 18:721-34. [PMID: 24479820 PMCID: PMC4000122 DOI: 10.1111/jcmm.12221] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 11/28/2013] [Indexed: 01/08/2023] Open
Abstract
Clinical complications associated with atherosclerotic plaques arise from luminal obstruction due to plaque growth or destabilization leading to rupture. Tumour necrosis factor ligand superfamily member 12 (TNFSF12) also known as TNF-related weak inducer of apoptosis (TWEAK) is a proinflammatory cytokine that participates in atherosclerotic plaque development, but its role in plaque stability remains unclear. Using two different approaches, genetic deletion of TNFSF12 and treatment with a TWEAK blocking mAb in atherosclerosis-prone mice, we have analysed the effect of TWEAK inhibition on atherosclerotic plaques progression and stability. Mice lacking both TNFSF12 and Apolipoprotein E (TNFSF12−/−ApoE−/−) exhibited a diminished atherosclerotic burden and lesion size in their aorta. Advanced atherosclerotic plaques of TNFSF12−/−ApoE−/− or anti-TWEAK treated mice exhibited an increase collagen/lipid and vascular smooth muscle cell/macrophage ratios compared with TNFSF12+/+ApoE−/− control mice, reflecting a more stable plaque phenotype. These changes are related with two different mechanisms, reduction of the inflammatory response (chemokines expression and secretion and nuclear factor kappa B activation) and decrease of metalloproteinase activity in atherosclerotic plaques of TNFSF12−/−ApoE−/−. A similar phenotype was observed with anti-TWEAK mAb treatment in TNFSF12+/+ApoE−/− mice. Brachiocephalic arteries were also examined since they exhibit additional features akin to human atherosclerotic plaques associated with instability and rupture. Features of greater plaque stability including augmented collagen/lipid ratio, reduced macrophage content, and less presence of lateral xanthomas, buried caps, medial erosion, intraplaque haemorrhage and calcium content were present in TNFSF12−/−ApoE−/− or anti-TWEAK treatment in TNFSF12+/+ApoE−/− mice. Overall, our data indicate that anti-TWEAK treatment has the capacity to diminish proinflamatory response associated with atherosclerotic plaque progression and to alter plaque morphology towards a stable phenotype.
Collapse
Affiliation(s)
- Cristina Sastre
- Lab. Patología Vascular, IIS-Fundación Jiménez Díaz, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Tajrishi MM, Zheng TS, Burkly LC, Kumar A. The TWEAK-Fn14 pathway: a potent regulator of skeletal muscle biology in health and disease. Cytokine Growth Factor Rev 2013; 25:215-25. [PMID: 24444596 DOI: 10.1016/j.cytogfr.2013.12.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Accepted: 12/15/2013] [Indexed: 12/24/2022]
Abstract
TNF-like weak inducer of apoptosis (TWEAK), a TNF superfamily ligand, and its bona fide receptor, the TNF receptor superfamily member fibroblast growth factor-inducible 14 (Fn14), represent a pivotal axis for shaping both physiological and pathological tissue responses to acute or chronic injury and disease. In recent years significant advances have been made in delineating the prominent role of TWEAK-Fn14 dyad in regulating skeletal muscle mass and metabolism. Also emerging from the broad study of tissue injury in skeletal muscle and other organs is the role of the TWEAK-Fn14 pathway in promoting fibrosis. This review article highlights recent advancements toward understanding how the TWEAK-Fn14 pathway regulates the response to various skeletal muscle insults and, more broadly, engages multiple mechanisms to drive tissue fibrosis.
Collapse
Affiliation(s)
- Marjan M Tajrishi
- Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, United States
| | - Timothy S Zheng
- Department of Immunology, Biogen Idec, 12 Cambridge Center, Cambridge, MA 02142, United States
| | - Linda C Burkly
- Department of Immunology, Biogen Idec, 12 Cambridge Center, Cambridge, MA 02142, United States.
| | - Ashok Kumar
- Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, United States.
| |
Collapse
|
47
|
Ucero AC, Benito-Martin A, Fuentes-Calvo I, Santamaria B, Blanco J, Lopez-Novoa JM, Ruiz-Ortega M, Egido J, Burkly LC, Martinez-Salgado C, Ortiz A. TNF-related weak inducer of apoptosis (TWEAK) promotes kidney fibrosis and Ras-dependent proliferation of cultured renal fibroblast. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1744-55. [PMID: 23748045 DOI: 10.1016/j.bbadis.2013.05.032] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Revised: 05/20/2013] [Accepted: 05/23/2013] [Indexed: 11/18/2022]
Abstract
Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) regulates apoptosis, proliferation and inflammation in renal epithelial cells and plays a role in acute kidney injury. However, there is little information on the chronic effects of TWEAK. We hypothesized that TWEAK may influence renal fibrosis and regulate kidney fibroblast biology, in part, through Ras pathway. We studied a chronic model of experimental unilateral ureteral obstruction in wild type and TWEAK deficient mice, and a murine model of systemic TWEAK overexpression. TWEAK actions were also explored in cultured renal and embryonic fibroblasts. TWEAK and TWEAK receptor expression was increased in the obstructed kidneys. The absence of TWEAK decreased early kidney tubular damage, inflammatory infiltrates and myofibroblast number. TWEAK deficient mice had decreased renal fibrosis 21days after obstruction, as assessed by extracellular matrix staining. In mice without prior underlying kidney disease, systemic overexpression of TWEAK induced kidney inflammation and fibrosis. In cultured fibroblasts, TWEAK induced proliferation through activation of the Ras/ERK pathway. TWEAK also activated nuclear factor κB (NFκB)-dependent inflammatory chemokine production in murine renal fibroblasts. In conclusion, lack of TWEAK reduces renal fibrosis in a model of persistent kidney insult and overexpression of TWEAK led to renal fibrosis. TWEAK actions on renal fibroblasts may contribute to the in vivo observations, as TWEAK promotes inflammatory activity and proliferation in fibroblast cultures.
Collapse
Affiliation(s)
- Alvaro C Ucero
- IIS-FundacionJimenezDiaz, Av. Reyes Católicos, Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Novoyatleva T, Schymura Y, Janssen W, Strobl F, Swiercz JM, Patra C, Posern G, Wietelmann A, Zheng TS, Schermuly RT, Engel FB. Deletion of Fn14 receptor protects from right heart fibrosis and dysfunction. Basic Res Cardiol 2013; 108:325. [PMID: 23325387 PMCID: PMC3597271 DOI: 10.1007/s00395-012-0325-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 11/29/2012] [Accepted: 12/17/2012] [Indexed: 12/14/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a fatal disease for which no cure is yet available. The leading cause of death in PAH is right ventricular (RV) failure. Previously, the TNF receptor superfamily member fibroblast growth factor-inducible molecule 14 (Fn14) has been associated with different fibrotic diseases. However, so far there is no study demonstrating a causal role for endogenous Fn14 signaling in RV or LV heart disease. The purpose of this study was to determine whether global ablation of Fn14 prevents RV fibrosis and remodeling improving heart function. Here, we provide evidence for a causative role of Fn14 in pulmonary artery banding (PAB)-induced RV fibrosis and dysfunction in mice. Fn14 expression was increased in the RV after PAB. Mice lacking Fn14 (Fn14−/−) displayed substantially reduced RV fibrosis and dysfunction following PAB compared to wild-type littermates. Cell culture experiments demonstrated that activation of Fn14 induces collagen expression via RhoA-dependent nuclear translocation of myocardin-related transcription factor-A (MRTF-A)/MAL. Furthermore, activation of Fn14 in vitro caused fibroblast proliferation and myofibroblast differentiation, which corresponds to suppression of PAB-induced RV fibrosis in Fn14−/− mice. Moreover, our findings suggest that Fn14 expression is regulated by endothelin-1 (ET-1) in cardiac fibroblasts. We conclude that Fn14 is an endogenous key regulator in cardiac fibrosis and suggest this receptor as potential new target for therapeutic interventions in heart failure.
Collapse
Affiliation(s)
- Tatyana Novoyatleva
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
The role of TWEAK/Fn14 in cardiac remodeling. Mol Biol Rep 2012; 39:9971-7. [PMID: 22752727 DOI: 10.1007/s11033-012-1867-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 06/19/2012] [Indexed: 10/28/2022]
Abstract
The pathophysiological basis of heart failure is cardiac remodeling, a process that comprises structural and functional changes including cardiomyocyte proliferation, hypertrophy, necrosis, apoptosis, autophagy, interstitial fibrosis, contractile dysfunction and ventricular dilatation. Accumulating evidence demonstrate that tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is involved in the process by binding its receptor fibroblast growth factor-inducible molecule 14 (Fn14). In this review, we will discuss the potential role of the TWEAK/Fn14 axis in cardiac remodeling, elucidate its possible mechanisms and explore new therapeutic targets for heart failure.
Collapse
|