1
|
Umbarkar P, Ejantkar S, Tousif S, Lal H. Mechanisms of Fibroblast Activation and Myocardial Fibrosis: Lessons Learned from FB-Specific Conditional Mouse Models. Cells 2021; 10:cells10092412. [PMID: 34572061 PMCID: PMC8471002 DOI: 10.3390/cells10092412] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 01/26/2023] Open
Abstract
Heart failure (HF) is a leading cause of morbidity and mortality across the world. Cardiac fibrosis is associated with HF progression. Fibrosis is characterized by the excessive accumulation of extracellular matrix components. This is a physiological response to tissue injury. However, uncontrolled fibrosis leads to adverse cardiac remodeling and contributes significantly to cardiac dysfunction. Fibroblasts (FBs) are the primary drivers of myocardial fibrosis. However, until recently, FBs were thought to play a secondary role in cardiac pathophysiology. This review article will present the evolving story of fibroblast biology and fibrosis in cardiac diseases, emphasizing their recent shift from a supporting to a leading role in our understanding of the pathogenesis of cardiac diseases. Indeed, this story only became possible because of the emergence of FB-specific mouse models. This study includes an update on the advancements in the generation of FB-specific mouse models. Regarding the underlying mechanisms of myocardial fibrosis, we will focus on the pathways that have been validated using FB-specific, in vivo mouse models. These pathways include the TGF-β/SMAD3, p38 MAPK, Wnt/β-Catenin, G-protein-coupled receptor kinase (GRK), and Hippo signaling. A better understanding of the mechanisms underlying fibroblast activation and fibrosis may provide a novel therapeutic target for the management of adverse fibrotic remodeling in the diseased heart.
Collapse
Affiliation(s)
- Prachi Umbarkar
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Correspondence: (P.U.); (H.L.); Tel.: +1-205-996-4248 (P.U.); +1-205-996-4219 (H.L.); Fax: +1-205-975-5104 (H.L.)
| | - Suma Ejantkar
- School of Health Professions, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Sultan Tousif
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Hind Lal
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Correspondence: (P.U.); (H.L.); Tel.: +1-205-996-4248 (P.U.); +1-205-996-4219 (H.L.); Fax: +1-205-975-5104 (H.L.)
| |
Collapse
|
2
|
Li Z, Zhang J, Wang M, Qiu F, Jin C, Fu G. Expression of farnesyl pyrophosphate synthase is increased in diabetic cardiomyopathy. Cell Biol Int 2021; 45:1393-1403. [PMID: 33595160 DOI: 10.1002/cbin.11573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/25/2021] [Accepted: 02/14/2021] [Indexed: 12/19/2022]
Abstract
Farnesyl pyrophosphate synthase (FPPS)-catalyzed isoprenoid intermediates are involved in diabetic cardiomyopathy. This study investigated the specific role of FPPS in the development of diabetic cardiomyopathy. We demonstrated that FPPS expression was elevated in both in vivo and in vitro models of diabetic cardiomyopathy. FPPS inhibition decreased the expression of proteins related to cardiac fibrosis and cardiomyocytic hypertrophy, including collagen I, collagen III, connective tissue growth factor, natriuretic factor, brain natriuretic peptide, and β-myosin heavy chain. Furthermore, FPPS inhibition and knockdown prevented phosphorylated c-Jun N-terminal kinase 1/2 (JNK1/2) activation in vitro. In addition, a JNK1/2 inhibitor downregulated high-glucose-induced responses to diabetic cardiomyopathy. Finally, immunofluorescence revealed that cardiomyocytic size was elevated by high glucose and was decreased by zoledronate, small-interfering farnesyl pyrophosphate synthase (siFPPS), and a JNK1/2 inhibitor. Taken together, our findings indicate that FPPS and JNK1/2 may be part of a signaling pathway that plays an important role in diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Zhengwei Li
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, PR China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang Province, PR China
| | - Jiefang Zhang
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, PR China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang Province, PR China
| | - Min Wang
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, PR China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang Province, PR China
| | - Fuyu Qiu
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, PR China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang Province, PR China
| | - Chongyin Jin
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, PR China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang Province, PR China
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, PR China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang Province, PR China
| |
Collapse
|
3
|
Li X, Garcia-Elias A, Benito B, Nattel S. The effects of cardiac stretch on atrial fibroblasts: Analysis of the evidence and potential role in atrial fibrillation. Cardiovasc Res 2021; 118:440-460. [PMID: 33576384 DOI: 10.1093/cvr/cvab035] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/27/2020] [Accepted: 02/09/2021] [Indexed: 01/06/2023] Open
Abstract
Atrial fibrillation (AF) is an important clinical problem. Chronic pressure/volume overload of the atria promotes AF, particularly via enhanced extracellular matrix (ECM) accumulation manifested as tissue fibrosis. Loading of cardiac cells causes cell-stretch that is generally considered to promote fibrosis by directly activating fibroblasts, the key cell-type responsible for ECM-production. The primary purpose of this article is to review the evidence regarding direct effects of stretch on cardiac fibroblasts, specifically: (i) the similarities and differences among studies in observed effects of stretch on cardiac-fibroblast function; (ii) the signaling-pathways implicated; and (iii) the factors that affect stretch-related phenotypes. Our review summarizes the most important findings and limitations in this area and gives an overview of clinical data and animal models related to cardiac stretch, with particular emphasis on the atria. We suggest that the evidence regarding direct fibroblast activation by stretch is weak and inconsistent, in part because of variability among studies in key experimental conditions that govern the results. Further work is needed to clarify whether, in fact, stretch induces direct activation of cardiac fibroblasts and if so, to elucidate the determining factors to ensure reproducible results. If mechanical load on fibroblasts proves not to be clearly profibrotic by direct actions, other mechanisms like paracrine influences, the effects of systemic mediators and/or the direct consequences of myocardial injury or death, might account for the link between cardiac stretch and fibrosis. Clarity in this area is needed to improve our understanding of AF pathophysiology and assist in therapeutic development.
Collapse
Affiliation(s)
- Xixiao Li
- Department of Medicine and Research Center, Montreal Heart Institute, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Anna Garcia-Elias
- Department of Medicine and Research Center, Montreal Heart Institute, Montreal, Canada
| | - Begoña Benito
- Vascular Biology and Metabolism Program, Vall d'Hebrón Research Institute (VHIR), Barcelona, Spain.,Cardiology Department, Hospital Universitari Vall d'Hebrón, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Stanley Nattel
- Department of Medicine and Research Center, Montreal Heart Institute, Montreal, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada.,Department of Pharmacology and Physiology of the Université de Montréal Faculty of Medicine, Montreal, Canada.,Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Essen, Germany.,IHU LIRYC and Fondation Bordeaux Université, Bordeaux, France
| |
Collapse
|
4
|
Xu Y, Rong J, Zhang Z. The emerging role of angiotensinogen in cardiovascular diseases. J Cell Physiol 2020; 236:68-78. [PMID: 32572956 DOI: 10.1002/jcp.29889] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022]
Abstract
Angiotensinogen (AGT) is the unique precursor of all angiotensin peptides. Many of the basic understandings of AGT in cardiovascular diseases have come from research efforts to define its effects on blood pressure regulation. The development of novel techniques targeting AGT manipulation such as genetic animal models, adeno-associated viral approaches, and antisense oligonucleotides made it possible to deeply investigate the relationship between AGT and cardiovascular diseases. In this brief review, we provide contemporary insights into the emerging role of AGT in cardiovascular diseases. In light of the recent progress, we emphasize some newly recognized features and mechanisms of AGT in heart failure, hypertension, atherosclerosis, and cardiovascular risk factors.
Collapse
Affiliation(s)
- Yinchuan Xu
- Department of Cardiology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jiabing Rong
- Department of Cardiology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zhaocai Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
5
|
Pasternak JA, MacPhee DJ, Harding JCS. Maternal and fetal thyroid dysfunction following porcine reproductive and respiratory syndrome virus2 infection. Vet Res 2020; 51:47. [PMID: 32228691 PMCID: PMC7106657 DOI: 10.1186/s13567-020-00772-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 03/15/2020] [Indexed: 12/20/2022] Open
Abstract
To better understand the host response to porcine reproductive and respiratory virus-2 (PRRSV2) we evaluated circulating thyroid hormone and associated gene expression in a late gestation challenge model. Pregnant gilts were inoculated at gestation day 85 and fetal samples collected at either 12 or 21 days post-infection (dpi). A subset of fetuses was selected for analysis based on viability and viral load categorized as either uninfected-viable (UNIF), high viral load viable (HV-VIA) or high viral load meconium stained (HV-MEC) and were compared with gestational age matched controls (CON). In dams, circulating levels of total T3 and T4 decreased in the acute period following infection and rebounded by 21 dpi. A similar effect was observed in fetuses, but was largely restricted to HV-VIA and HV-MEC, with minimal decrease noted in UNIF relative to CON at 21 dpi. Gene expression in fetal heart at 12 dpi showed significant decompensatory transcription of thyroid hormone transporters (SLC16A2) and deiodinases (DIO2, DIO3), which was not observed in brain. Correspondingly, genes associated with cell cycle progression (CDK1,2,4) were downregulated in only the heart of highly infected fetuses, while expression of their inhibitor (CDKN1A) was upregulated in both tissues. Finally, expression of genes associated with cardiac stress including CAMKD and AGT were upregulated in the hearts of highly infected fetuses, and a shift in expression of MYH6 to MYH7 was observed in HV-MEC fetuses specifically. Collectively, the results suggest PRRSV2 infection causes a hypothyroid state that disproportionally impacts the fetal heart over the brain.
Collapse
Affiliation(s)
- J Alex Pasternak
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Dr, Saskatoon, SK, S7N 5B4, Canada. .,Department of Animal Sciences, Purdue University, 270 S. Russell St, West Lafayette, IN, 47907, USA.
| | - Daniel J MacPhee
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Dr, Saskatoon, SK, S7N 5B4, Canada
| | - John C S Harding
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Dr, Saskatoon, SK, S7N 5B4, Canada
| |
Collapse
|
6
|
Zeglinski MR, Moghadam AR, Ande SR, Sheikholeslami K, Mokarram P, Sepehri Z, Rokni H, Mohtaram NK, Poorebrahim M, Masoom A, Toback M, Sareen N, Saravanan S, Jassal DS, Hashemi M, Marzban H, Schaafsma D, Singal P, Wigle JT, Czubryt MP, Akbari M, Dixon IM, Ghavami S, Gordon JW, Dhingra S. Myocardial Cell Signaling During the Transition to Heart Failure. Compr Physiol 2018; 9:75-125. [DOI: 10.1002/cphy.c170053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
7
|
Bageghni SA, Hemmings KE, Zava N, Denton CP, Porter KE, Ainscough JFX, Drinkhill MJ, Turner NA. Cardiac fibroblast-specific p38α MAP kinase promotes cardiac hypertrophy via a putative paracrine interleukin-6 signaling mechanism. FASEB J 2018; 32:4941-4954. [PMID: 29601781 PMCID: PMC6629170 DOI: 10.1096/fj.201701455rr] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/26/2018] [Indexed: 12/11/2022]
Abstract
Recent studies suggest that cardiac fibroblast-specific p38α MAPK contributes to the development of cardiac hypertrophy, but the underlying mechanism is unknown. Our study used a novel fibroblast-specific, tamoxifen-inducible p38α knockout (KO) mouse line to characterize the role of fibroblast p38α in modulating cardiac hypertrophy, and we elucidated the mechanism. Myocardial injury was induced in tamoxifen-treated Cre-positive p38α KO mice or control littermates via chronic infusion of the β-adrenergic receptor agonist isoproterenol. Cardiac function was assessed by pressure-volume conductance catheter analysis and was evaluated for cardiac hypertrophy at tissue, cellular, and molecular levels. Isoproterenol infusion in control mice promoted overt cardiac hypertrophy and dysfunction (reduced ejection fraction, increased end systolic volume, increased cardiac weight index, increased cardiomyocyte area, increased fibrosis, and up-regulation of myocyte fetal genes and hypertrophy-associated microRNAs). Fibroblast-specific p38α KO mice exhibited marked protection against myocardial injury, with isoproterenol-induced alterations in cardiac function, histology, and molecular markers all being attenuated. In vitro mechanistic studies determined that cardiac fibroblasts responded to damaged myocardium by secreting several paracrine factors known to induce cardiomyocyte hypertrophy, including IL-6, whose secretion was dependent upon p38α activity. In conclusion, cardiac fibroblast p38α contributes to cardiomyocyte hypertrophy and cardiac dysfunction, potentially via a mechanism involving paracrine fibroblast-to-myocyte IL-6 signaling.-Bageghni, S. A., Hemmings, K. E., Zava, N., Denton, C. P., Porter, K. E., Ainscough, J. F. X., Drinkhill, M. J., Turner, N. A. Cardiac fibroblast-specific p38α MAP kinase promotes cardiac hypertrophy via a putative paracrine interleukin-6 signaling mechanism.
Collapse
Affiliation(s)
- Sumia A. Bageghni
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, United Kingdom; and
| | - Karen E. Hemmings
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, United Kingdom; and
| | - Ngonidzashe Zava
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, United Kingdom; and
| | - Christopher P. Denton
- Centre for Rheumatology, Division of Medicine, University College London, London, United Kingdom
| | - Karen E. Porter
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, United Kingdom; and
| | - Justin F. X. Ainscough
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, United Kingdom; and
| | - Mark J. Drinkhill
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, United Kingdom; and
| | - Neil A. Turner
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, United Kingdom; and
| |
Collapse
|
8
|
Cardiomyocyte-specific deletion of GSK-3β leads to cardiac dysfunction in a diet induced obesity model. Int J Cardiol 2018; 259:145-152. [PMID: 29398139 DOI: 10.1016/j.ijcard.2018.01.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/19/2017] [Accepted: 01/02/2018] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND RATIONALE Obesity, an independent risk factor for the development of myocardial diseases is a growing healthcare problem worldwide. It's well established that GSK-3β is critical to cardiac pathophysiology. However, the role cardiomyocyte (CM) GSK-3β in diet-induced cardiac dysfunction is unknown. METHODS CM-specific GSK-3β knockout (CM-GSK-3β-KO) and littermate controls (WT) mice were fed either a control diet (CD) or high-fat diet (HFD) for 55weeks. Cardiac function was assessed by transthoracic echocardiography. RESULTS At baseline, body weights and cardiac function were comparable between the WT and CM-GSK-3β-KOs. However, HFD-fed CM-GSK-3β-KO mice developed severe cardiac dysfunction. Consistently, both heart weight/tibia length and lung weight/tibia length were significantly elevated in the HFD-fed CM-GSK-3β-KO mice. The impaired cardiac function and adverse ventricular remodeling in the CM-GSK-3β-KOs were independent of body weight or the lean/fat mass composition as HFD-fed CM-GSK-3β-KO and controls demonstrated comparable body weight and body masses. At the molecular level, on a CD, CM-GSK-3α compensated for the loss of CM-GSK-3β, as evident by significantly reduced GSK-3αs21 phosphorylation (activation) resulting in a preserved canonical β-catenin ubiquitination pathway and cardiac function. However, this protective compensatory mechanism is lost with HFD, leading to excessive accumulation of β-catenin in HFD-fed CM-GSK-3β-KO hearts, resulting in adverse ventricular remodeling and cardiac dysfunction. CONCLUSION In summary, these results suggest that cardiac GSK-3β is crucial to protect against obesity-induced adverse ventricular remodeling and cardiac dysfunction.
Collapse
|
9
|
Gerena Y, Lozada JG, Collazo BJ, Méndez-Álvarez J, Méndez-Estrada J, De Mello WC. Losartan counteracts the effects of cardiomyocyte swelling on glucose uptake and insulin receptor substrate-1 levels. Peptides 2017; 96:38-43. [PMID: 28889965 PMCID: PMC5618797 DOI: 10.1016/j.peptides.2017.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 09/01/2017] [Accepted: 09/05/2017] [Indexed: 11/20/2022]
Abstract
A growing body of evidence demonstrates an association between Angiotensin II (Ang II) receptor blockers (ARBs) and enhanced glucose metabolism during ischemic heart disease. Despite these encouraging results, the mechanisms responsible for these effects during ischemia remain poorly understood. In this study we investigated the influence of losartan, an AT1 receptor blocker, and secreted Ang II (sAng II) on glucose uptake and insulin receptor substrate (IRS-1) levels during cardiomyocyte swelling. H9c2 cells were differentiated to cardiac muscle and the levels of myogenin, Myosin Light Chain (MLC), and membrane AT1 receptors were measured using flow cytometry. Intracellular Ang II (iAng II) was overexpressed in differentiated cardiomyocytes and swelling was induced after incubation with hypotonic solution for 40min. Glucose uptake and IRS-1 levels were monitored by flow cytometry using 2-NBDG fluorescent glucose (10μM) or an anti-IRS-1 monoclonal antibody in the presence or absence of losartan (10-7M). Secreted Angiotensin II was quantified from the medium using a specific Ang II-EIA kit. To evaluate the relationship between sAng II and losartan effects on glucose uptake, transfected cells were pretreated with the drug for 24h and then exposed to hypotonic solution in the presence or absence of the secreted peptide. The results indicate that: (1) swelling of transfected cardiomyocytes decreased glucose uptake and induced the secretion of Ang II to the extracellular medium; (2) losartan antagonized the effects of swelling on glucose uptake and IRS-1 levels in transfected cardiomyocytes; (3) the effects of losartan on glucose uptake were observed during swelling only in the presence of sAng II in the culture medium. Our study demonstrates that both losartan and sAng II have essential roles in glucose metabolism during cardiomyocyte swelling.
Collapse
Affiliation(s)
- Yamil Gerena
- Department of Pharmacology and Toxicology, School of Medicine, University of Puerto Rico, Medical Sciences Campus, P.O. Box 365067, San Juan 00936-5067, Puerto Rico.
| | - Janice Griselle Lozada
- Department of Pharmacology and Toxicology, School of Medicine, University of Puerto Rico, Medical Sciences Campus, P.O. Box 365067, San Juan 00936-5067, Puerto Rico.
| | - Bryan Jael Collazo
- Department of Pharmacology and Toxicology, School of Medicine, University of Puerto Rico, Medical Sciences Campus, P.O. Box 365067, San Juan 00936-5067, Puerto Rico.
| | - Jarold Méndez-Álvarez
- Department of Pharmacology and Toxicology, School of Medicine, University of Puerto Rico, Medical Sciences Campus, P.O. Box 365067, San Juan 00936-5067, Puerto Rico.
| | - Jennifer Méndez-Estrada
- Department of Pharmacology and Toxicology, School of Medicine, University of Puerto Rico, Medical Sciences Campus, P.O. Box 365067, San Juan 00936-5067, Puerto Rico.
| | - Walmor C De Mello
- Department of Pharmacology and Toxicology, School of Medicine, University of Puerto Rico, Medical Sciences Campus, P.O. Box 365067, San Juan 00936-5067, Puerto Rico.
| |
Collapse
|
10
|
Ontiveros-Torres MÁ, Labra-Barrios ML, Díaz-Cintra S, Aguilar-Vázquez AR, Moreno-Campuzano S, Flores-Rodríguez P, Luna-Herrera C, Mena R, Perry G, Florán-Garduño B, Luna-Muñoz J, Luna-Arias JP. Fibrillar Amyloid-β Accumulation Triggers an Inflammatory Mechanism Leading to Hyperphosphorylation of the Carboxyl-Terminal End of Tau Polypeptide in the Hippocampal Formation of the 3×Tg-AD Transgenic Mouse. J Alzheimers Dis 2017; 52:243-69. [PMID: 27031470 DOI: 10.3233/jad-150837] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is a degenerative and irreversible disorder whose progressiveness is dependent on age. It is histopathologically characterized by the massive accumulation of insoluble forms of tau and amyloid-β (Aβ) asneurofibrillary tangles and neuritic plaques, respectively. Many studies have documented that these two polypeptides suffer several posttranslational modifications employing postmortem tissue sections from brains of patients with AD. In order to elucidate the molecular mechanisms underlying the posttranslational modifications of key players in this disease, including Aβ and tau, several transgenic mouse models have been developed. One of these models is the 3×Tg-AD transgenic mouse, carrying three transgenes encoding APPSWE, S1M146V, and TauP301L proteins. To further characterize this transgenicmouse, we determined the accumulation of fibrillar Aβ as a function of age in relation to the hyperphosphorylation patterns of TauP301L at both its N- and C-terminus in the hippocampal formation by immunofluorescence and confocal microscopy. Moreover, we searched for the expression of activated protein kinases and mediators of inflammation by western blot of wholeprotein extracts from hippocampal tissue sections since 3 to 28 months as well. Our results indicate that the presence of fibrillar Aβ deposits correlates with a significant activation of astrocytes and microglia in subiculum and CA1 regions of hippocampus. Accordingly, we also observed a significant increase in the expression of TNF-α associated to neuritic plaques and glial cells. Importantly, there is an overexpression of the stress activated protein kinases SAPK/JNK and Cdk-5 in pyramidal neurons, which might phosphorylate several residues at the C-terminus of TauP301L. Therefore, the accumulation of Aβ oligomers results in an inflammatory environment that upregulates kinases involved in hyperphosphorylation of TauP301L polypeptide.
Collapse
Affiliation(s)
- Miguel Ángel Ontiveros-Torres
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Col. San Pedro Zacatenco, Ciudad de México, México
| | - María Luisa Labra-Barrios
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Col. San Pedro Zacatenco, Ciudad de México, México
| | - Sofía Díaz-Cintra
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Juriquilla, Querétaro, Qro., México
| | | | - Samadhi Moreno-Campuzano
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Col. San Pedro Zacatenco, Ciudad de México, México
| | - Paola Flores-Rodríguez
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Col. San Pedro Zacatenco, Ciudad de México, México.,Present address: Departamento de Fisiología, Facultad de Medicina y Nutrición, Universidad Juárez del Estado de Durango, Durango, Dgo., México
| | - Claudia Luna-Herrera
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Campus Zacatenco, Ciudad de México, México
| | - Raúl Mena
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Col. San Pedro Zacatenco, Ciudad de México, México
| | - George Perry
- College of Sciences, University of Texas at San Antonio, San Antonio, TX, USA
| | - Benjamín Florán-Garduño
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Col. San Pedro Zacatenco, Ciudad de México, México
| | - José Luna-Muñoz
- Banco Nacional de Cerebros, Laboratorio Nacional de Servicios Experimentales, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Col. San Pedro Zacatenco, Ciudad de México, México
| | - Juan Pedro Luna-Arias
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Col. San Pedro Zacatenco, Ciudad de México, México
| |
Collapse
|
11
|
Abstract
In this chapter, we discuss the manner through which the immune system regulates the cardiovascular system in health and disease. We define the cardiovascular system and elements of atherosclerotic disease, the main focus in this chapter. Herein we elaborate on the disease process that can result in myocardial infarction (heart attack), ischaemic stroke and peripheral arterial disease. We have discussed broadly the homeostatic mechanisms in place that help autoregulate the cardiovascular system including the vital role of cholesterol and lipid clearance as well as the role lipid homeostasis plays in cardiovascular disease in the context of atherosclerosis. We then elaborate on the role played by the immune system in this setting, namely, major players from the innate and adaptive immune system, as well as discussing in greater detail specifically the role played by monocytes and macrophages.This chapter should represent an overview of the role played by the immune system in cardiovascular homeostasis; however further reading of the references cited can expand the reader's knowledge of the detail, and we point readers to many excellent reviews which summarise individual immune systems and their role in cardiovascular disease.
Collapse
Affiliation(s)
- Mohammed Shamim Rahman
- Division of Immunology and Inflammation, Department of Medicine, Imperial College London, London, UK
| | - Kevin Woollard
- Division of Immunology and Inflammation, Department of Medicine, Imperial College London, London, UK.
| |
Collapse
|
12
|
Stoppel WL, Kaplan DL, Black LD. Electrical and mechanical stimulation of cardiac cells and tissue constructs. Adv Drug Deliv Rev 2016; 96:135-55. [PMID: 26232525 DOI: 10.1016/j.addr.2015.07.009] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 07/16/2015] [Accepted: 07/25/2015] [Indexed: 12/19/2022]
Abstract
The field of cardiac tissue engineering has made significant strides over the last few decades, highlighted by the development of human cell derived constructs that have shown increasing functional maturity over time, particularly using bioreactor systems to stimulate the constructs. However, the functionality of these tissues is still unable to match that of native cardiac tissue and many of the stem-cell derived cardiomyocytes display an immature, fetal like phenotype. In this review, we seek to elucidate the biological underpinnings of both mechanical and electrical signaling, as identified via studies related to cardiac development and those related to an evaluation of cardiac disease progression. Next, we review the different types of bioreactors developed to individually deliver electrical and mechanical stimulation to cardiomyocytes in vitro in both two and three-dimensional tissue platforms. Reactors and culture conditions that promote functional cardiomyogenesis in vitro are also highlighted. We then cover the more recent work in the development of bioreactors that combine electrical and mechanical stimulation in order to mimic the complex signaling environment present in vivo. We conclude by offering our impressions on the important next steps for physiologically relevant mechanical and electrical stimulation of cardiac cells and engineered tissue in vitro.
Collapse
|
13
|
Feng H, Gerilechaogetu F, Golden HB, Nizamutdinov D, Foster DM, Glaser SS, Dostal DE. p38α MAPK inhibits stretch-induced JNK activation in cardiac myocytes through MKP-1. Int J Cardiol 2016; 203:145-55. [DOI: 10.1016/j.ijcard.2015.10.109] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 09/20/2015] [Accepted: 10/12/2015] [Indexed: 01/18/2023]
|
14
|
Kishore R, Krishnamurthy P, Garikipati VNS, Benedict C, Nickoloff E, Khan M, Johnson J, Gumpert AM, Koch WJ, Verma SK. Interleukin-10 inhibits chronic angiotensin II-induced pathological autophagy. J Mol Cell Cardiol 2015; 89:203-13. [PMID: 26549357 PMCID: PMC4689660 DOI: 10.1016/j.yjmcc.2015.11.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/13/2015] [Accepted: 11/02/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND Although autophagy is an essential cellular salvage process to maintain cellular homeostasis, pathological autophagy can lead to cardiac abnormalities and ultimately heart failure. Therefore, a tight regulation of autophagic process would be important to treat chronic heart failure. Previously, we have shown that IL-10 strongly inhibited pressure overload-induced hypertrophy and heart failure, but role of IL-10 in regulation of pathological autophagy is unknown. Here we tested the hypothesis that IL-10 inhibits angiotensin II-induced pathological autophagy and this process, in part, leads to improve cardiac function. METHODS AND RESULTS Chronic Ang II strongly induced mortality, cardiac dysfunction in IL-10 Knockout mice. IL-10 deletion exaggerated pathological autophagy in response to Ang II treatment. In isolated cardiac myocytes, IL-10 attenuated Ang II-induced pathological autophagy and activated Akt/mTORC1 signaling. Pharmacological or molecular inhibition of Akt and mTORC1 signaling attenuated IL-10 effects on Ang II-induced pathological autophagy. Furthermore, lysosomal inhibition in autophagic flux experiments further confirmed that IL-10 inhibits pathological autophagy via mTORC1 signaling. CONCLUSION Our data demonstrate a novel role of IL-10 in regulation of pathological autophagy; thus can act as a potential therapeutic molecule for treatment of chronic heart disease.
Collapse
Affiliation(s)
- Raj Kishore
- Center for Translational Medicine, Temple University, Philadelphia 19140, USA; Department of Pharmacology, Temple University, Philadelphia 19140, USA.
| | - Prasanna Krishnamurthy
- Department of Cardiovascular Science, Houston Methodist Research Institute, Houston 77030, USA
| | | | - Cindy Benedict
- Center for Translational Medicine, Temple University, Philadelphia 19140, USA
| | - Emily Nickoloff
- Center for Translational Medicine, Temple University, Philadelphia 19140, USA
| | - Mohsin Khan
- Center for Translational Medicine, Temple University, Philadelphia 19140, USA
| | - Jennifer Johnson
- Center for Translational Medicine, Temple University, Philadelphia 19140, USA
| | - Anna M Gumpert
- Center for Translational Medicine, Temple University, Philadelphia 19140, USA
| | - Walter J Koch
- Center for Translational Medicine, Temple University, Philadelphia 19140, USA; Department of Pharmacology, Temple University, Philadelphia 19140, USA
| | - Suresh Kumar Verma
- Center for Translational Medicine, Temple University, Philadelphia 19140, USA.
| |
Collapse
|
15
|
Dostal D, Glaser S, Baudino TA. Cardiac Fibroblast Physiology and Pathology. Compr Physiol 2015; 5:887-909. [DOI: 10.1002/cphy.c140053] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
16
|
Qi Y, Zhang K, Wu Y, Xu Z, Yong QC, Kumar R, Baker KM, Zhu Q, Chen S, Guo S. Novel mechanism of blood pressure regulation by forkhead box class O1-mediated transcriptional control of hepatic angiotensinogen. Hypertension 2014; 64:1131-40. [PMID: 25069665 DOI: 10.1161/hypertensionaha.114.03970] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The renin-angiotensin system is a major determinant of blood pressure regulation. It consists of a cascade of enzymatic reactions involving 3 components: angiotensinogen, renin, and angiotensin-converting enzyme, which generate angiotensin II as a biologically active product. Angiotensinogen is largely produced in the liver, acting as a major determinant of the circulating renin-angiotensin system, which exerts acute hemodynamic effects on blood pressure regulation. How the expression of angiotensinogen is regulated is not completely understood. Here, we hypothesize that angiotensinogen is regulated by forkhead transcription factor forkhead box class O1 (Foxo1), an insulin-suppressed transcription factor, and thereby controls blood pressure in mice. We generated liver-specific Foxo1 knockout mice, which exhibited a reduction in plasma angiotensinogen and angiotensin II levels and a significant decrease in blood pressure. Using hepatocyte cultures, we demonstrated that overexpression of Foxo1 increased angiotensinogen expression, whereas hepatocytes lacking Foxo1 demonstrated a reduction of angiotensinogen gene expression and partially impaired insulin inhibition on angiotensinogen gene expression. Furthermore, mouse angiotensinogen prompter analysis demonstrated that the angiotensinogen promoter region contains a functional Foxo1-binding site, which is responsible for both Foxo1 stimulation and insulin suppression on the promoter activity. Together, these data demonstrate that Foxo1 regulates hepatic angiotensinogen gene expression and controls plasma angiotensinogen and angiotensin II levels, modulating blood pressure control in mice.
Collapse
Affiliation(s)
- Yajuan Qi
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.)
| | - Kebin Zhang
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.)
| | - Yuxin Wu
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.)
| | - Zihui Xu
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.)
| | - Qian Chen Yong
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.)
| | - Rajesh Kumar
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.)
| | - Kenneth M Baker
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.)
| | - Qinglei Zhu
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.)
| | - Shouwen Chen
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.)
| | - Shaodong Guo
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.).
| |
Collapse
|
17
|
Wagner MA, Siddiqui MAQ. The JAK-STAT pathway in hypertrophic stress signaling and genomic stress response. JAKSTAT 2014; 1:131-41. [PMID: 24058762 PMCID: PMC3670293 DOI: 10.4161/jkst.20702] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The JAK-STAT signaling pathway plays a central role in transducing stress and growth signals in the hypertrophic heart. Unlike most signal transducers, JAKs and STATs signal in a number of different ways, both within the JAK-STAT pathway and in collaboration with other signaling pathways. In this review, we discuss how IL-6 activates cells lacking IL-6 receptors through trans-signaling and examine JAK-STAT pathway interaction with GPCR-linked pathways both within and between cells. Finally, we discuss recent studies showing how the JAK-STAT pathway can intersect with a general transcriptional regulatory mechanism to effect transcription of STAT-dependent stress response genes.
Collapse
Affiliation(s)
- Michael A Wagner
- Department of Cell Biology; Center for Cardiovascular and Muscle Research; State University of New York Downstate Medical Center; Brooklyn, NY USA
| | | |
Collapse
|
18
|
Feng H, Gerilechaogetu F, Golden HB, Nizamutdinov D, Foster DM, Glaser S, Dostal DE. Paracrine communication between mechanically stretched myocytes and fibroblasts. Methods Mol Biol 2014; 1066:57-66. [PMID: 23955733 DOI: 10.1007/978-1-62703-604-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Mechanical stretch is a major factor for myocardial hypertrophy and heart failure. Stretch activates mechanical sensors from cardiac myocytes, leading to a series of signal transduction cascades, which can result in cell malfunction and remodeling. It is well known that mechanical stretch also induces the release of paracrine factors from cardiac fibroblasts, as well as myocytes. Due to complicated circumstance of heart tissue, it is difficult to fully investigate the characteristics of these factors in situ. Here we describe static stretch and conditioned medium experiments as methods to examine the function of paracrine factors between primary cultured cardiac myocytes and fibroblasts.
Collapse
Affiliation(s)
- Hao Feng
- Texas A&M Health Science Center, College of Medicine, Cardiovascular Research Institute, Temple, TX, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Dostal DE, Feng H, Nizamutdinov D, Golden HB, Afroze SH, Dostal JD, Jacob JC, Foster DM, Tong C, Glaser S, Gerilechaogetu F. Mechanosensing and Regulation of Cardiac Function. ACTA ACUST UNITED AC 2014; 5:314. [PMID: 25485172 PMCID: PMC4255974 DOI: 10.4172/2155-9880.1000314] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The role of mechanical force as an important regulator of structure and function of mammalian cells, tissues, and organs has recently been recognized. However, mechanical overload is a pathogenesis or comorbidity existing in a variety of heart diseases, such as hypertension, aortic regurgitation and myocardial infarction. Physical stimuli sensed by cells are transmitted through intracellular signal transduction pathways resulting in altered physiological responses or pathological conditions. Emerging evidence from experimental studies indicate that β1-integrin and the angiotensin II type I (AT1) receptor play critical roles as mechanosensors in the regulation of heart contraction, growth and leading to heart failure. Integrin link the extracellular matrix and the intracellular cytoskeleton to initiate the mechanical signalling, whereas, the AT1 receptor could be activated by mechanical stress through an angiotensin-II-independent mechanism. Recent studies show that both Integrin and AT1 receptor and their downstream signalling factors including MAPKs, AKT, FAK, ILK and GTPase regulate heart function in cardiac myocytes. In this review we describe the role of mechanical sensors residing within the plasma membrane, mechanical sensor induced downstream signalling factors and its potential roles in cardiac contraction and growth.
Collapse
Affiliation(s)
- David E Dostal
- Central Texas Veterans Health Care System, Temple, Texas, USA ; Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - Hao Feng
- Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - Damir Nizamutdinov
- Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - Honey B Golden
- Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - Syeda H Afroze
- Scott & White Healthcare - Digestive Disease Research Centre, Temple, Texas, USA
| | - Joseph D Dostal
- Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - John C Jacob
- Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - Donald M Foster
- Central Texas Veterans Health Care System, Temple, Texas, USA
| | - Carl Tong
- Systems Biology and Translational Medicine, the Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| | - Shannon Glaser
- Central Texas Veterans Health Care System, Temple, Texas, USA ; Scott & White Healthcare - Digestive Disease Research Centre, Temple, Texas, USA
| | - Fnu Gerilechaogetu
- Division of Molecular Cardiology, Cardiovascular Research Institute, Texas A&M University Health Science Centre, College of Medicine, Temple, Texas, USA
| |
Collapse
|
20
|
Ye F, Yuan F, Li X, Cooper N, Tinney JP, Keller BB. Gene expression profiles in engineered cardiac tissues respond to mechanical loading and inhibition of tyrosine kinases. Physiol Rep 2013; 1:e00078. [PMID: 24303162 PMCID: PMC3841024 DOI: 10.1002/phy2.78] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 08/07/2013] [Indexed: 12/17/2022] Open
Abstract
Engineered cardiac tissues (ECTs) are platforms to investigate cardiomyocyte maturation and functional integration, the feasibility of generating tissues for cardiac repair, and as models for pharmacology and toxicology bioassays. ECTs rapidly mature in vitro to acquire the features of functional cardiac muscle and respond to mechanical load with increased proliferation and maturation. ECTs are now being investigated as platforms for in vitro models for human diseases and for pharmacologic screening for drug toxicities. We tested the hypothesis that global ECT gene expression patterns are complex and sensitive to mechanical loading and tyrosine kinase inhibitors similar to the maturing myocardium. We generated ECTs from day 14.5 rat embryo ventricular cells, as previously published, and then conditioned constructs after 5 days in culture for 48 h with mechanical stretch (5%, 0.5 Hz) and/or the p38 MAPK (p38 mitogen-activated protein kinase) inhibitor BIRB796. RNA was isolated from individual ECTs and assayed using a standard Agilent rat 4 × 44k V3 microarray and Pathway Analysis software for transcript expression fold changes and changes in regulatory molecules and networks. Changes in expression were confirmed by quantitative-polymerase chain reaction (q-PCR) for selected regulatory molecules. At the threshold of a 1.5-fold change in expression, stretch altered 1559 transcripts, versus 1411 for BIRB796, and 1846 for stretch plus BIRB796. As anticipated, top pathways altered in response to these stimuli include cellular development, cellular growth and proliferation; tissue development; cell death, cell signaling, and small molecule biochemistry as well as numerous other pathways. Thus, ECTs display a broad spectrum of altered gene expression in response to mechanical load and/or tyrosine kinase inhibition, reflecting a complex regulation of proliferation, differentiation, and architectural alignment of cardiomyocytes and noncardiomyocytes within ECT.
Collapse
Affiliation(s)
- Fei Ye
- Kosair Charities Pediatric Heart Research Program, Cardiovascular Innovation Institute, University of Louisville Louisville, Kentucky ; Affiliated Hospital of Guiyang Medical College Guiyang, China
| | | | | | | | | | | |
Collapse
|
21
|
Anthrax lethal toxin induces acute diastolic dysfunction in rats through disruption of the phospholamban signaling network. Int J Cardiol 2013; 168:3884-95. [PMID: 23907041 DOI: 10.1016/j.ijcard.2013.06.050] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 05/09/2013] [Accepted: 06/28/2013] [Indexed: 01/08/2023]
Abstract
BACKGROUND Anthrax lethal toxin (LT), secreted by Bacillus anthracis, causes severe cardiac dysfunction by unknown mechanisms. LT specifically cleaves the docking domains of MAPKK (MEKs); thus, we hypothesized that LT directly impairs cardiac function through dysregulation of MAPK signaling mechanisms. METHODS AND RESULTS In a time-course study of LT toxicity, echocardiography revealed acute diastolic heart failure accompanied by pulmonary regurgitation and left atrial dilation in adult Sprague-Dawley rats at time points corresponding to dysregulated JNK, phospholamban (PLB) and protein phosphatase 2A (PP2A) myocardial signaling. Using isolated rat ventricular myocytes, we identified the MEK7-JNK1-PP2A-PLB signaling axis to be important for regulation of intracellular calcium (Ca(2+)(i)) handling, PP2A activation and targeting of PP2A-B56α to Ca(2+)(i) handling proteins, such as PLB. Through a combination of gain-of-function and loss-of-function studies, we demonstrated that over-expression of MEK7 protects against LT-induced PP2A activation and Ca(2+)(i) dysregulation through activation of JNK1. Moreover, targeted phosphorylation of PLB-Thr(17) by Akt improved sarcoplasmic reticulum Ca(2+)(i) release and reuptake during LT toxicity. Co-immunoprecipitation experiments further revealed the pivotal role of MEK7-JNK-Akt complex formation for phosphorylation of PLB-Thr(17) during acute LT toxicity. CONCLUSIONS Our findings support a cardiogenic mechanism of LT-induced diastolic dysfunction, by which LT disrupts JNK1 signaling and results in Ca(2+)(i) dysregulation through diminished phosphorylation of PLB by Akt and increased dephosphorylation of PLB by PP2A. Integration of the MEK7-JNK1 signaling module with Akt represents an important stress-activated signalosome that may confer protection to sustain cardiac contractility and maintain normal levels of Ca(2+)(i) through PLB-T(17) phosphorylation.
Collapse
|
22
|
Lal H, Verma SK, Feng H, Golden HB, Gerilechaogetu F, Nizamutdinov D, Foster DM, Glaser SS, Dostal DE. Caveolin and β1-integrin coordinate angiotensinogen expression in cardiac myocytes. Int J Cardiol 2012; 168:436-45. [PMID: 23058350 DOI: 10.1016/j.ijcard.2012.09.131] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 06/23/2012] [Accepted: 09/22/2012] [Indexed: 12/20/2022]
Abstract
BACKGROUND The cardiac renin-angiotensin system (RAS) has been implicated in mediating myocyte hypertrophy and remodeling, although the biochemical mechanisms responsible for regulating the local RAS are poorly understood. Caveolin-1 (Cav-1)/Cav-3 double-knockout mice display cardiac hypertrophy, and in vitro disruption of lipid rafts/caveolae using methyl-β-cyclodextrin (MβCD) abolishes cardiac protection. METHODS In this study, neonatal rat ventricular myocytes (NRVM) were used to determine whether lipid rafts/caveolae may be involved in the regulation of angiotensinogen (Ao) gene expression, a substrate of the RAS system. RESULTS Treatment with MβCD caused a time-dependent upregulation of Ao gene expression, which was associated with differential regulation of mitogen-activated protein (MAP) kinases ERK1/2, p38 and JNK phosphorylation. JNK was highly phosphorylated shortly after MβCD treatment (2-30 min), whereas marked activation of ERK1/2 and p38 occurred much later (2-4h). β1D-Integrin was required for MβCD-induced activation of the MAP kinases. Pharmacologic inhibition of ERK1/2 and JNK enhanced MβCD-induced Ao gene expression, whereas p38 blockade inhibited this response. Adenovirus-mediated expression of wild-type p38α enhanced MβCD-induced Ao gene expression; conversely expression of dominant negative p38α blocked the stimulatory effects of MβCD. Expression of Cav-3 siRNA stimulated Ao gene expression, whereas overexpression of Cav-3 was inhibitory. Cav-1 and Cav-3 expression levels were found to be positively regulated by p38, but unaffected by ERK1/2 and JNK. CONCLUSION Collectively, these studies indicate that lipid rafts/caveolae couple to Ao gene expression through a mechanism that involves β1-integrin and the differential actions of MAP kinase family members.
Collapse
Affiliation(s)
- Hind Lal
- Center for Translational Medicine, Temple University, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Verma SK, Krishnamurthy P, Barefield D, Singh N, Gupta R, Lambers E, Thal M, Mackie A, Hoxha E, Ramirez V, Qin G, Sadayappan S, Ghosh AK, Kishore R. Interleukin-10 treatment attenuates pressure overload-induced hypertrophic remodeling and improves heart function via signal transducers and activators of transcription 3-dependent inhibition of nuclear factor-κB. Circulation 2012; 126:418-29. [PMID: 22705886 DOI: 10.1161/circulationaha.112.112185] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Inflammation plays a critical role in adverse cardiac remodeling and heart failure. Therefore, approaches geared toward inhibiting inflammation may provide therapeutic benefits. We tested the hypotheses that genetic deletion of interleukin-10 (IL-10), a potent antiinflammatory cytokine, exacerbates pressure overload-induced adverse cardiac remodeling and hypertrophy and that IL-10 therapy inhibits this pathology. METHODS AND RESULTS Cardiac hypertrophy was induced in wild-type and IL-10 knockout mice by isoproterenol (ISO) infusion. ISO-induced left ventricular dysfunction and hypertrophic remodeling, including fibrosis and fetal gene expression, were further exaggerated in knockout mice compared with wild-type mice. Systemic recombinant mouse IL-10 administration markedly improved left ventricular function and not only inhibited but also reversed ISO-induced cardiac remodeling. Intriguingly, a very similar cardioprotective response of IL-10 was found in transverse aortic constriction-induced hypertrophy and heart failure models. In neonatal rat ventricular myocytes and H9c2 myoblasts, ISO activated nuclear factor-κB and inhibited signal transducers and activators of transcription 3 (STAT3) phosphorylation. Interestingly, IL-10 suppressed ISO-induced nuclear factor-κB activation and attenuated STAT3 inhibition. Moreover, pharmacological and genetic inhibition of STAT3 reversed the protective effects of IL-10, whereas ectopic expression of constitutively active STAT3 mimicked the IL-10 responses on the ISO effects, confirming that the IL-10-mediated inhibition of nuclear factor-κB is STAT3 dependent. CONCLUSION Taken together, our results suggest IL-10 treatment as a potential therapeutic approach to limit the progression of pressure overload-induced adverse cardiac remodeling.
Collapse
Affiliation(s)
- Suresh Kumar Verma
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, 303 E Superior St, Chicago IL 60611, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Chang C, Zhang C, Zhao X, Kuang X, Tang H, Xiao X. Differential regulation of mitogen-activated protein kinase signaling pathways in human with different types of mitral valvular disease. J Surg Res 2012; 181:49-59. [PMID: 22664133 DOI: 10.1016/j.jss.2012.05.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 04/11/2012] [Accepted: 05/08/2012] [Indexed: 02/05/2023]
Abstract
BACKGROUND Mitogen-activated protein kinases (MAPKs) are considered to play a prominent role in cardiac development, function, and pathogenesis. The different types of mitral valvular disease (MVD), including mitral regurgitation (MR) and mitral stenosis (MS), have different underlying pathophysiologic changes, but the precise intracellular signal transduction mechanisms are not clear. Thus, we investigated the differential regulation of MAPK signaling pathways in humans with different types of MVD. METHODS Left atrial appendage tissue samples from 32 patients with MVD who were undergoing mitral valve replacement surgery were studied. Serum angiotensin II concentrations were measured using enzyme-linked immunosorbent assay. The expression of MAPK pathway-related genes and proteins was assessed using quantitative polymerase chain reaction, Western blot, and immunohistochemistry. RESULTS Echocardiography showed that patients with MS had a greater left atrial pressure overload than those with MR. The relative amounts of angiotensin II, extracellular signal-regulated kinase 1, p38α, c-Jun N-terminal kinase 2, c-Fos, activating transcription factor 2, and c-Jun mRNA were significantly upregulated in those with MS compared with those with MR (P < 0.05). The serum angiotensin II concentrations were significantly increased in those with MS compared with those with MR (P = 0.017). Substantial changes in the phosphorylated forms of the MAPK proteins were detected. Phosphorylated extracellular signal-regulated kinase 1/2, and phosphorylated p38 were significantly increased in those with MS compared with those with MR (P < 0.001), and phosphorylated c-Jun N-terminal kinase in the MR group was significantly greater than that in the MS group (P < 0.001). Histologically, more serious myocardial cells losses, myolysis, and interstitial fibrosis were detected in the MS group. CONCLUSIONS The different types of MVD have different hemodynamic characteristics, and different MAPK pathways were activated in the MR and MS groups, which could lead to diverse left atrial histologic changes.
Collapse
Affiliation(s)
- Chao Chang
- Department of Thoracic and Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China
| | | | | | | | | | | |
Collapse
|
25
|
Tsai CT, Chiang FT, Tseng CD, Yu CC, Wang YC, Lai LP, Hwang JJ, Lin JL. Mechanical Stretch of Atrial Myocyte Monolayer Decreases Sarcoplasmic Reticulum Calcium Adenosine Triphosphatase Expression and Increases Susceptibility to Repolarization Alternans. J Am Coll Cardiol 2011; 58:2106-15. [DOI: 10.1016/j.jacc.2011.07.039] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 07/05/2011] [Accepted: 07/12/2011] [Indexed: 11/25/2022]
|
26
|
Turner NA. Therapeutic regulation of cardiac fibroblast function: targeting stress-activated protein kinase pathways. Future Cardiol 2011; 7:673-91. [DOI: 10.2217/fca.11.41] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
27
|
Verma SK, Lal H, Golden HB, Gerilechaogetu F, Smith M, Guleria RS, Foster DM, Lu G, Dostal DE. Rac1 and RhoA differentially regulate angiotensinogen gene expression in stretched cardiac fibroblasts. Cardiovasc Res 2010; 90:88-96. [PMID: 21131638 DOI: 10.1093/cvr/cvq385] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
AIMS Angiotensin II (Ang II) stimulates cardiac remodelling and fibrosis in the mechanically overloaded myocardium. Although Rho GTPases regulate several cellular processes, including myocardial remodelling, involvement in mediating mechanical stretch-induced regulation of angiotensinogen (Ao), the precursor to Ang II, remains to be determined. We, therefore, examined the role and associated signalling mechanisms of Rho GTPases (Rac1 and RhoA) in regulation of Ao gene expression in a stretch model of neonatal rat cardiac fibroblasts (CFs). METHODS AND RESULTS CFs were plated on deformable stretch membranes. Equiaxial mechanical stretch caused significant activation of both Rac1 and RhoA within 2-5 min. Rac1 activity returned to control levels after 4 h, whereas RhoA remained at a high level of activity until the end of the stretch period (24 h). Mechanical stretch initially caused a moderate decrease in Ao gene expression, but was significantly increased at 8-24 h. RhoA had a major role in mediating both the stretch-induced inhibition of Ao at 4 h and the subsequent upregulation of Ao expression at 24 h. β₁ integrin receptor blockade by Tac β₁ expression impaired acute (2 and 15 min) stretch-induced Rac1 activation, but increased RhoA activity. Molecular experiments revealed that Ao gene expression was inhibited by Rac1 through both JNK-dependent and independent mechanisms, and stimulated by RhoA through a p38-dependent mechanism. CONCLUSION These results indicate that stretch-induced activation of Rac1 and RhoA differentially regulates Ao gene expression by modulating p38 and JNK activation.
Collapse
Affiliation(s)
- Suresh K Verma
- Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Cardiac fibroblasts: at the heart of myocardial remodeling. Pharmacol Ther 2009; 123:255-78. [PMID: 19460403 DOI: 10.1016/j.pharmthera.2009.05.002] [Citation(s) in RCA: 763] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Accepted: 05/05/2009] [Indexed: 12/24/2022]
Abstract
Cardiac fibroblasts are the most prevalent cell type in the heart and play a key role in regulating normal myocardial function and in the adverse myocardial remodeling that occurs with hypertension, myocardial infarction and heart failure. Many of the functional effects of cardiac fibroblasts are mediated through differentiation to a myofibroblast phenotype that expresses contractile proteins and exhibits increased migratory, proliferative and secretory properties. Cardiac myofibroblasts respond to proinflammatory cytokines (e.g. TNFalpha, IL-1, IL-6, TGF-beta), vasoactive peptides (e.g. angiotensin II, endothelin-1, natriuretic peptides) and hormones (e.g. noradrenaline), the levels of which are increased in the remodeling heart. Their function is also modulated by mechanical stretch and changes in oxygen availability (e.g. ischaemia-reperfusion). Myofibroblast responses to such stimuli include changes in cell proliferation, cell migration, extracellular matrix metabolism and secretion of various bioactive molecules including cytokines, vasoactive peptides and growth factors. Several classes of commonly prescribed therapeutic agents for cardiovascular disease also exert pleiotropic effects on cardiac fibroblasts that may explain some of their beneficial outcomes on the remodeling heart. These include drugs for reducing hypertension (ACE inhibitors, angiotensin receptor blockers, beta-blockers), cholesterol levels (statins, fibrates) and insulin resistance (thiazolidinediones). In this review, we provide insight into the properties of cardiac fibroblasts that underscores their importance in the remodeling heart, including their origin, electrophysiological properties, role in matrix metabolism, functional responses to environmental stimuli and ability to secrete bioactive molecules. We also review the evidence suggesting that certain cardiovascular drugs can reduce myocardial remodeling specifically via modulatory effects on cardiac fibroblasts.
Collapse
|