1
|
Senescent cardiac fibroblasts: A key role in cardiac fibrosis. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166642. [PMID: 36669578 DOI: 10.1016/j.bbadis.2023.166642] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/19/2023]
Abstract
Cardiac fibroblasts are a cell population that controls the homeostasis of the extracellular matrix and orchestrates a damage response to maintain cardiac architecture and performance. Due to these functions, fibroblasts play a central role in cardiac fibrosis development, and there are large differences in matrix protein secretion profiles between fibroblasts from aged versus young animals. Senescence is a multifactorial and complex process that has been associated with inflammatory and fibrotic responses. After damage, transient cellular senescence is usually beneficial, as these cells promote tissue repair. However, the persistent presence of senescent cells within a tissue is linked with fibrosis development and organ dysfunction, leading to aging-related diseases such as cardiovascular pathologies. In the heart, early cardiac fibroblast senescence after myocardial infarction seems to be protective to avoid excessive fibrosis; however, in non-infarcted models of cardiac fibrosis, cardiac fibroblast senescence has been shown to be deleterious. Today, two new classes of drugs, termed senolytics and senostatics, which eliminate senescent cells or modify senescence-associated secretory phenotype, respectively, arise as novel therapeutical strategies to treat aging-related pathologies. However, further studies will be needed to evaluate the extent of the utility of senotherapeutic drugs in cardiac diseases, in which pathological context and temporality of the intervention must be considered.
Collapse
|
2
|
Zavadakova A, Vistejnova L, Tonarova P. Functional responses of dermal fibroblasts to low nutrition and pro-inflammatory stimuli mimicking a wound environment in vitro. In Vitro Cell Dev Biol Anim 2022; 58:643-657. [PMID: 35948856 DOI: 10.1007/s11626-022-00713-7] [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: 03/24/2022] [Accepted: 07/16/2022] [Indexed: 11/30/2022]
Abstract
Dermal fibroblasts (DF) constitute one of key cells involved in wound healing. However, the functions they perform in wound conditions remain poorly understood. This study involved exposing DF to low nutrition and to low nutrition + LPS for 5 d as conditions representing the wound. Although DF exhibited increasing metabolic activity in time under all conditions including control, the proliferation did not change in both low nutrition and low nutrition + LPS. Only the low nutrition + LPS was found to potentiate the migration and pro-inflammatory phenotype (IL6 release) of DF. The potential of DF to contract collagen hydrogel declined only under low nutrition as a consequence of low cell number. The expression of α-SMA was reduced under both conditions independently of the cell number. The remodeling capability of DF was affected under both conditions as documented by the enhanced MMP2 activity. Finally, the production of collagen type I was not affected by either condition. The study shows that low nutrition as the single factor is able to delay the healing process. Moreover, the addition of the mild pro-inflammatory stimulus represented by LPS may amplify the cell response in case of decreased α-SMA expression or excite DF to produce IL6 impairing the healing process.
Collapse
Affiliation(s)
- Anna Zavadakova
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, Pilsen, Czech Republic.
| | - Lucie Vistejnova
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, Pilsen, Czech Republic.,Department of Histology and Embryology, Faculty of Medicine in Pilsen, Charles University, Karlovarska 48, Pilsen, Czech Republic
| | - Pavla Tonarova
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, Pilsen, Czech Republic.,Institute of Pathological Physiology, 1st Faculty of Medicine, Charles University, U Nemocnice 5, Prague, Czech Republic
| |
Collapse
|
3
|
Espitia-Corredor JA, Boza P, Espinoza-Pérez C, Lillo JM, Rimassa-Taré C, Machuca V, Osorio-Sandoval JM, Vivar R, Bolivar S, Pardo-Jiménez V, Sánchez-Ferrer CF, Peiró C, Díaz-Araya G. Angiotensin II Triggers NLRP3 Inflammasome Activation by a Ca 2+ Signaling-Dependent Pathway in Rat Cardiac Fibroblast Ang-II by a Ca 2+-Dependent Mechanism Triggers NLRP3 Inflammasome in CF. Inflammation 2022; 45:2498-2512. [PMID: 35867264 DOI: 10.1007/s10753-022-01707-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/13/2022] [Accepted: 06/09/2022] [Indexed: 11/05/2022]
Abstract
Angiotensin II (Ang-II) is a widely studied hypertensive, profibrotic, and pro-inflammatory peptide. In the heart, cardiac fibroblasts (CF) express type 1 angiotensin II receptors (AT1R), Toll-like receptor-4 (TLR4), and the NLRP3 inflammasome complex, which play important roles in pro-inflammatory processes. When activated, the NLRP3 inflammasome triggers proteolytic cleavage of pro-IL-1, resulting in its activation. However, in CF the mechanism by which Ang-II assembles and activates the NLRP3 inflammasome remains not fully known. To elucidate this important point, we stimulated TLR4 receptors in CF and evaluated the signaling pathways by which Ang-II triggers the assembly and activity. In cultured rat CF, pro-IL-1β levels, NLRP3, ASC, and caspase-1 expression levels were determined by Western blot. NLRP3 inflammasome complex assembly was analyzed by immunocytochemistry, whereas by ELISA, we analyzed NLRP3 inflammasome activity and [Formula: see text] release. In CF, Ang-II triggered NLRP3 inflammasome assembly and caspase-1 activity; and in LPS-pretreated CF, Ang-II also triggered [Formula: see text] secretion. These effects were blocked by losartan (AT1R antagonist), U73221 (PLC inhibitor), 2-APB (IP3R antagonist), and BAPTA-AM (Ca2+ chelator) indicating that the AT1R/PLC/IP3R/Ca2+ pathway is involved. Finally, bafilomycin A1 prevented Ang-II-induced [Formula: see text] secretion, indicating that a non-classical protein secretion mechanism is involved. These findings suggest that in CF, Ang-II by a Ca2+-dependent mechanism triggers NLRP3 inflammasome assembly and activation leading to [Formula: see text] secretion through a non-conventional protein secretion mechanism.
Collapse
Affiliation(s)
- Jenaro Antonio Espitia-Corredor
- Laboratory of Molecular Pharmacology, Faculty of Chemical and Pharmaceutical Sciences, Department of Pharmacological & Toxicological Chemistry, University of Chile, Santiago, Chile.,Faculty of Medicine, Department of Pharmacology, Universidad Autónoma de Madrid, Madrid, Spain.,PhD Programme in Pharmacology and Physiology, Doctoral School, Universidad Autónoma de Madrid, Madrid, Spain.,Faculty of Chemical and Pharmaceutical Sciences, Advanced Center of Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile
| | - Pía Boza
- Laboratory of Molecular Pharmacology, Faculty of Chemical and Pharmaceutical Sciences, Department of Pharmacological & Toxicological Chemistry, University of Chile, Santiago, Chile
| | - Claudio Espinoza-Pérez
- Laboratory of Molecular Pharmacology, Faculty of Chemical and Pharmaceutical Sciences, Department of Pharmacological & Toxicological Chemistry, University of Chile, Santiago, Chile
| | - José Miguel Lillo
- Laboratory of Molecular Pharmacology, Faculty of Chemical and Pharmaceutical Sciences, Department of Pharmacological & Toxicological Chemistry, University of Chile, Santiago, Chile
| | - Constanza Rimassa-Taré
- Laboratory of Molecular Pharmacology, Faculty of Chemical and Pharmaceutical Sciences, Department of Pharmacological & Toxicological Chemistry, University of Chile, Santiago, Chile
| | - Víctor Machuca
- Laboratory of Molecular Pharmacology, Faculty of Chemical and Pharmaceutical Sciences, Department of Pharmacological & Toxicological Chemistry, University of Chile, Santiago, Chile
| | - José Miguel Osorio-Sandoval
- Laboratory of Molecular Pharmacology, Faculty of Chemical and Pharmaceutical Sciences, Department of Pharmacological & Toxicological Chemistry, University of Chile, Santiago, Chile
| | - Raúl Vivar
- Molecular and Clinical Pharmacology Program, Faculty of Medicine, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago, Chile
| | - Samir Bolivar
- Faculty of Chemistry and Pharmacy, Universidad del Atlántico, Barranquilla, Colombia
| | - Viviana Pardo-Jiménez
- Laboratory of Molecular Pharmacology, Faculty of Chemical and Pharmaceutical Sciences, Department of Pharmacological & Toxicological Chemistry, University of Chile, Santiago, Chile
| | - Carlos Félix Sánchez-Ferrer
- Faculty of Medicine, Department of Pharmacology, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigaciones Sanitarias (IdiPAZ), Madrid, Spain
| | - Concepción Peiró
- Faculty of Medicine, Department of Pharmacology, Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigaciones Sanitarias (IdiPAZ), Madrid, Spain
| | - Guillermo Díaz-Araya
- Laboratory of Molecular Pharmacology, Faculty of Chemical and Pharmaceutical Sciences, Department of Pharmacological & Toxicological Chemistry, University of Chile, Santiago, Chile. .,Faculty of Chemical and Pharmaceutical Sciences, Advanced Center of Chronic Diseases (ACCDiS), University of Chile, Santiago, Chile.
| |
Collapse
|
4
|
Gut Microbiome and Organ Fibrosis. Nutrients 2022; 14:nu14020352. [PMID: 35057530 PMCID: PMC8781069 DOI: 10.3390/nu14020352] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 02/07/2023] Open
Abstract
Fibrosis is a pathological process associated with most chronic inflammatory diseases. It is defined by an excessive deposition of extracellular matrix proteins and can affect nearly every tissue and organ system in the body. Fibroproliferative diseases, such as intestinal fibrosis, liver cirrhosis, progressive kidney disease and cardiovascular disease, often lead to severe organ damage and are a leading cause of morbidity and mortality worldwide, for which there are currently no effective therapies available. In the past decade, a growing body of evidence has highlighted the gut microbiome as a major player in the regulation of the innate and adaptive immune system, with severe implications in the pathogenesis of multiple immune-mediated disorders. Gut microbiota dysbiosis has been associated with the development and progression of fibrotic processes in various organs and is predicted to be a potential therapeutic target for fibrosis management. In this review we summarize the state of the art concerning the crosstalk between intestinal microbiota and organ fibrosis, address the relevance of diet in different fibrotic diseases and discuss gut microbiome-targeted therapeutic approaches that are current being explored.
Collapse
|
5
|
Parra-Flores P, Espitia-Corredor J, Espinoza-Pérez C, Queirolo C, Ayala P, Brüggendieck F, Salas-Hernández A, Pardo-Jiménez V, Díaz-Araya G. Toll-Like Receptor 4 Activation Prevents Rat Cardiac Fibroblast Death Induced by Simulated Ischemia/Reperfusion. Front Cardiovasc Med 2021; 8:660197. [PMID: 34169098 PMCID: PMC8217466 DOI: 10.3389/fcvm.2021.660197] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/13/2021] [Indexed: 01/04/2023] Open
Abstract
Death of cardiac fibroblasts (CFs) by ischemia/reperfusion (I/R) has major implications for cardiac wound healing. In in vivo models of myocardial infarction, toll-like receptor 4 (TLR4) activation has been reported as a cardioprotector; however, it remains unknown whether TLR4 activation can prevent CF death triggered by simulated I/R (sI/R). In this study, we analyzed TLR4 activation in neonate CFs exposed to an in vitro model of sI/R and explored the participation of the pro-survival kinases Akt and ERK1/2. Simulated ischemia was performed in a free oxygen chamber in an ischemic medium, whereas reperfusion was carried out in normal culture conditions. Cell viability was analyzed by trypan blue exclusion and the MTT assay. Necrotic and apoptotic cell populations were evaluated by flow cytometry. Protein levels of phosphorylated forms of Akt and ERK1/2 were analyzed by Western blot. We showed that sI/R triggers CF death by necrosis and apoptosis. In CFs exposed only to simulated ischemia or only to sI/R, blockade of the TLR4 with TAK-242 further reduced cell viability and the activation of Akt and ERK1/2. Preconditioning with lipopolysaccharide (LPS) or treatment with LPS in ischemia or reperfusion was not protective. However, LPS incubation during both ischemia and reperfusion periods prevented CF viability loss induced by sI/R. Furthermore, LPS treatment reduced the sub-G1 population, but not necrosis of CFs exposed to sI/R. On the other hand, the protective effects exhibited by LPS were abolished when TLR4 was blocked and Akt and ERK1/2 were inhibited. In conclusion, our results suggest that TLR4 activation protects CFs from apoptosis induced by sI/R through the activation of Akt and ERK1/2 signaling pathways.
Collapse
Affiliation(s)
- Pablo Parra-Flores
- Laboratorio de Farmacología Molecular, Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Jenaro Espitia-Corredor
- Laboratorio de Farmacología Molecular, Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile.,Department of Pharmacology, Faculty of Medicine, Instituto de Investigación Sanitaria Hospital Universitario La Paz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Claudio Espinoza-Pérez
- Laboratorio de Farmacología Molecular, Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Cristian Queirolo
- Laboratorio de Farmacología Molecular, Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Pedro Ayala
- Departamento de Enfermedades Respiratorias, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisca Brüggendieck
- Laboratorio de Farmacología Molecular, Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Aimee Salas-Hernández
- Laboratorio de Farmacología Molecular, Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile.,Department of Pharmacology, Toxicology and Pharmacodependence, Pharmacy Faculty, University of Costa Rica, San José, Costa Rica
| | - Viviana Pardo-Jiménez
- Laboratorio de Farmacología Molecular, Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Guillermo Díaz-Araya
- Laboratorio de Farmacología Molecular, Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| |
Collapse
|
6
|
Salas-Hernández A, Espinoza-Pérez C, Vivar R, Espitia-Corredor J, Lillo J, Parra-Flores P, Sánchez-Ferrer CF, Peiró C, Díaz-Araya G. Resolvin D1 and E1 promote resolution of inflammation in rat cardiac fibroblast in vitro. Mol Biol Rep 2021; 48:57-66. [PMID: 33459958 DOI: 10.1007/s11033-020-06133-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 12/24/2020] [Indexed: 12/16/2022]
Abstract
Cardiac fibroblasts (CFs) have a key role in the inflammatory response after cardiac injury and are necessary for wound healing. Resolvins are potent agonists that control the duration and magnitude of inflammation. They decrease mediators of pro-inflammatory expression, reduce neutrophil migration to inflammation sites, promote the removal of microbes and apoptotic cells, and reduce exudate. However, whether resolvins can prevent pro-inflammatory-dependent effects in CFs is unknown. Thus, the present work was addressed to study whether resolvin D1 and E1 (RvD1 and RvE1) can prevent pro-inflammatory effects on CFs after lipopolysaccharide (LPS) challenge. For this, CFs were stimulated with LPS, in the presence or absence of RvD1 or RvE1, to analyze its effects on intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion protein 1 (VCAM-1), monocyte adhesion and the cytokine levels of tumor necrosis factor alpha (TNF-α), interleukin-6(IL-6), interleukin-1beta (IL-1β), monocyte chemoattractant protein-1 (MCP-1) and interleukin-10 (IL-10). Our results showed that CFs are expressing ALX/FPR2 and ChemR23, RvD1 and RvE1 receptors, respectively. RvD1 and RvE1 prevent the increase of ICAM-1 and VCAM-1 protein levels and the adhesion of spleen mononuclear cells to CFs induced by LPS. Finally, RvD1, but not RvE1, prevents the LPS-induced increase of IL-6, MCP-1, TNF-α, and IL-10. In conclusion, our findings provide evidence that in CFs, RvD1 and RvE1 might actively participate in the prevention of inflammatory response triggered by LPS.
Collapse
Affiliation(s)
- Aimeé Salas-Hernández
- Department of Chemical Pharmacology and Toxicology, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santos Dumont 964, Independencia, Santiago, Chile.,Department of Pharmacology, Toxicology and Pharmacodependence, Pharmacy Faculty, University of Costa Rica, San José, Costa Rica
| | - Claudio Espinoza-Pérez
- Department of Chemical Pharmacology and Toxicology, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santos Dumont 964, Independencia, Santiago, Chile
| | - Raúl Vivar
- Pharmacology Program, Biomedical Sciences Institute, University of Chile, Independencia 1027, Independencia, Santiago, Chile
| | - Jenaro Espitia-Corredor
- Department of Chemical Pharmacology and Toxicology, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santos Dumont 964, Independencia, Santiago, Chile
| | - José Lillo
- Department of Chemical Pharmacology and Toxicology, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santos Dumont 964, Independencia, Santiago, Chile
| | - Pablo Parra-Flores
- Department of Chemical Pharmacology and Toxicology, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santos Dumont 964, Independencia, Santiago, Chile
| | - Carlos F Sánchez-Ferrer
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid and Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Concepción Peiró
- Department of Pharmacology, Faculty of Medicine, Universidad Autónoma de Madrid and Instituto de Investigación Sanitaria Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Guillermo Díaz-Araya
- Department of Chemical Pharmacology and Toxicology, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santos Dumont 964, Independencia, Santiago, Chile. .,Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, University of Chile, Santos Dumont 964, Independencia, Santiago, Chile.
| |
Collapse
|
7
|
Xiao Z, Kong B, Yang H, Dai C, Fang J, Qin T, Huang H. Key Player in Cardiac Hypertrophy, Emphasizing the Role of Toll-Like Receptor 4. Front Cardiovasc Med 2020; 7:579036. [PMID: 33324685 PMCID: PMC7725871 DOI: 10.3389/fcvm.2020.579036] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/02/2020] [Indexed: 12/20/2022] Open
Abstract
Toll-like receptor 4 (TLR4), a key pattern recognition receptor, initiates the innate immune response and leads to chronic and acute inflammation. In the past decades, accumulating evidence has implicated TLR4-mediated inflammatory response in regulation of myocardium hypertrophic remodeling, indicating that regulation of the TLR4 signaling pathway may be an effective strategy for managing cardiac hypertrophy's pathophysiology. Given TLR4's significance, it is imperative to review the molecular mechanisms and roles underlying TLR4 signaling in cardiac hypertrophy. Here, we comprehensively review the current knowledge of TLR4-mediated inflammatory response and its interaction ligands and co-receptors, as well as activation of various intracellular signaling. We also describe the associated roles in promoting immune cell infiltration and inflammatory mediator secretion, that ultimately cause cardiac hypertrophy. Finally, we provide examples of some of the most promising drugs and new technologies that have the potential to attenuate TLR4-mediated inflammatory response and prevent or reverse the ominous cardiac hypertrophy outcomes.
Collapse
Affiliation(s)
- Zheng Xiao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Bin Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Hongjie Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Chang Dai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jin Fang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Tianyou Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
8
|
Rikard SM, Athey TL, Nelson AR, Christiansen SLM, Lee JJ, Holmes JW, Peirce SM, Saucerman JJ. Multiscale Coupling of an Agent-Based Model of Tissue Fibrosis and a Logic-Based Model of Intracellular Signaling. Front Physiol 2019; 10:1481. [PMID: 31920691 PMCID: PMC6928129 DOI: 10.3389/fphys.2019.01481] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 11/18/2019] [Indexed: 12/14/2022] Open
Abstract
Wound healing and fibrosis following myocardial infarction (MI) is a dynamic process involving many cell types, extracellular matrix (ECM), and inflammatory cues. As both incidence and survival rates for MI increase, management of post-MI recovery and associated complications are an increasingly important focus. Complexity of the wound healing process and the need for improved therapeutics necessitate a better understanding of the biochemical cues that drive fibrosis. To study the progression of cardiac fibrosis across spatial and temporal scales, we developed a novel hybrid multiscale model that couples a logic-based differential equation (LDE) model of the fibroblast intracellular signaling network with an agent-based model (ABM) of multi-cellular tissue remodeling. The ABM computes information about cytokine and growth factor levels in the environment including TGFβ, TNFα, IL-1β, and IL-6, which are passed as inputs to the LDE model. The LDE model then computes the network signaling state of individual cardiac fibroblasts within the ABM. Based on the current network state, fibroblasts make decisions regarding cytokine secretion and deposition and degradation of collagen. Simulated fibroblasts respond dynamically to rapidly changing extracellular environments and contribute to spatial heterogeneity in model predicted fibrosis, which is governed by many parameters including cell density, cell migration speeds, and cytokine levels. Verification tests confirmed that predictions of the coupled model and network model alone were consistent in response to constant cytokine inputs and furthermore, a subset of coupled model predictions were validated with in vitro experiments with human cardiac fibroblasts. This multiscale framework for cardiac fibrosis will allow for systematic screening of the effects of molecular perturbations in fibroblast signaling on tissue-scale extracellular matrix composition and organization.
Collapse
Affiliation(s)
- S Michaela Rikard
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Thomas L Athey
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Anders R Nelson
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States
| | - Steven L M Christiansen
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Jia-Jye Lee
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Jeffrey W Holmes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States.,Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
9
|
Zhang S, Jia X, Zhang Q, Zhang L, Yang J, Hu C, Shi J, Jiang X, Lu J, Shen H. Neutrophil extracellular traps activate lung fibroblast to induce polymyositis-related interstitial lung diseases via TLR9-miR-7-Smad2 pathway. J Cell Mol Med 2019; 24:1658-1669. [PMID: 31821687 PMCID: PMC6991674 DOI: 10.1111/jcmm.14858] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 06/18/2019] [Accepted: 07/23/2019] [Indexed: 12/13/2022] Open
Abstract
Excessive neutrophil extracellular trap (NET) formation may contribute to polymyositis (PM)‐associated interstitial lung diseases (ILD), but the underlying mechanism is not fully revealed. In this study, we found that NET accelerated the progression of ILD and promoted pulmonary fibrosis (PF) in vivo. miR‐7 expression was down‐regulated in lung tissue of PM group than control group, and NETs further decreased miR‐7 expression. TLR9 and Smad2 were up‐regulated in lung tissue of PM group than control group, and NETs further increased TLR9 and Smad2 expressions. In vitro experiments showed that PMA‐treated NETs accelerated the proliferation of LF and their differentiation into myofibroblast (MF), whereas DNase I decreased the promotion effect of NETs. Neutrophil extracellular trap components myeloperoxidase (MPO) and histone 3 also promoted the proliferation and differentiation of LF. In addition, we demonstrated that TLR9 involved in the regulation of NETs on LF proliferation and differentiation, and confirmed the interaction between miR‐7 and Smad2 in LF. Finally, miR‐7‐Smad2 pathway was confirmed to be involved in the regulation of TLR9 on LF proliferation and differentiation. Therefore, NETs promote PM‐related ILD, and TLR9‐miR‐7‐Smad2 signalling pathway is involved in the proliferation of LFs and their differentiation into MFs.
Collapse
Affiliation(s)
- Sigong Zhang
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
| | - Xueqin Jia
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
| | - Qiuyue Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Li Zhang
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
| | - Jing Yang
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
| | - Caihong Hu
- Department of Endocrinology, Lanzhou University Second Hospital, Lanzhou, China
| | - Junnian Shi
- Department of Pneumology, Lanzhou University Second Hospital, Lanzhou, China
| | - Xiao Jiang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jinyue Lu
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
| | - Haili Shen
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
10
|
Lappas M. Identification of SMAD3 as a Novel Mediator of Inflammation in Human Myometrium In Vitro. Mediators Inflamm 2018; 2018:3140420. [PMID: 30363688 PMCID: PMC6180979 DOI: 10.1155/2018/3140420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 07/31/2018] [Accepted: 08/09/2018] [Indexed: 02/01/2023] Open
Abstract
Preterm birth remains the primary cause of early neonatal death and is a major determinant for long-term health consequences. Aberrant intrauterine inflammation and infection are known to augment the synthesis of proinflammatory cytokines and induce uterine contractions, which can subsequently lead to preterm birth. The transforming growth factor-β (TGF-β) superfamily members regulate numerous cellular processes through the activation of intracellular mediators known as mothers against decapentaplegic homolog (SMADs). Studies in nongestational tissues have shown that SMAD3 plays a role in immune regulation and inflammation; however, its role in human labour remains unknown. Thus, the present study aimed at (i) characterising the expression of SMAD3 in the human myometrium; (ii) determining the effect of bacterial and viral products and proinflammatory cytokines on SMAD3 transcriptional activity in primary human myometrial cells; and (iii) investigating the effect of SMAD3 siRNA knockdown on the production of prolabour mediators in primary human myometrial cells. Phosphorylated (i.e., active) SMAD3 protein expression was lower in the myometrium after spontaneous term labour compared to the myometrium from nonlabouring women. Using a luciferase assay, the proinflammatory cytokines IL-1β and TNF, and viral analogue polyinosinic : polycytidylic acid (poly(I : C)) significantly reduced SMAD3 transcriptional activity in human primary myometrial cells. Loss-of-function studies found that SMAD3 knockdown in myometrial cells significantly increased IL-1β- and poly(I : C)-induced proinflammatory cytokines (IL-1A, IL-6), chemokines (IL-8, MCP-1), the adhesion molecule ICAM-1, COX-2 mRNA expression, and subsequent PGF2α release. In conclusion, SMAD3 deficiency is associated with increased production of proinflammatory and prolabour mediators in the human myometrium.
Collapse
Affiliation(s)
- Martha Lappas
- Obstetrics, Nutrition, and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
- Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| |
Collapse
|
11
|
Hu J, Shi B, Liu X, Jiang M, Yuan C, Jiang B, Song Y, Zeng Y, Wang G. The activation of Toll-like receptor 4 reverses tumor differentiation in human glioma U251 cells via Notch pathway. Int Immunopharmacol 2018; 64:33-41. [PMID: 30145468 DOI: 10.1016/j.intimp.2018.08.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 08/05/2018] [Accepted: 08/15/2018] [Indexed: 12/23/2022]
Abstract
Toll-like receptors (TLRs) are closely related to cancer. However, the mechanism for TLR regulation of cancer is not fully understood. Our previous studies demonstrated that toll-like receptor (TLR) 4 functions to maintain the un-differential stem cell phenotypes of human endothelial progenitor cells. In this study, we found that human glioma cells expressed several TLRs. The activation of TLR4 by LPS in glioma U251 cells induced the expression of cytokines, including IL-1β, IL-6, IL-8, and TNFα, suggesting the functional expression of TLR4. Nude mouse in vivo studies showed that LPS treatment promoted tumor growth, and decreased mouse survival. But LPS treatment did not promote tumor cell proliferation in vitro. Meanwhile, we found that LPS treatment down-regulated the expression of glial fibrillary acidic protein (GFAP), an important differentiation maker of glioma, at both mRNA and protein levels. TLR4 activation also down-regulated GFAP in glioma Hs683 cells. LPS did not induce the activation of MAPKs, but induced the activation of NF-κB. However, pharmacological inhibition of NF-κB signaling did not reverse the down-regulation of GFAP. Furthermore, we found that LPS induced the activation of Notch pathway, which was MyD88-dependent, and Notch inhibition reversed the down-regulation of GFAP. In addition, LPS treatment up-regulated stem cell makers, including CD34 and CD133. Taken together, these results suggested that in human glioma U251 cells, TLR4 functions to reverse tumor differentiation, and it may be a target for glioma prevention and therapy.
Collapse
Affiliation(s)
- Jinyue Hu
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan 410004, China.
| | - Bizhi Shi
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China
| | - Xueting Liu
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan 410004, China
| | - Manli Jiang
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan 410004, China
| | - Chuang Yuan
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan 410004, China
| | - Binyuan Jiang
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan 410004, China
| | - Yinghui Song
- Department of Oncology, Changsha Central Hospital, Changsha, Hunan 410004, China; Changsha Cancer Institute, Changsha Central Hospital, Changsha, Hunan 410004, China
| | - Yanhua Zeng
- Department of Oncology, Changsha Central Hospital, Changsha, Hunan 410004, China; Changsha Cancer Institute, Changsha Central Hospital, Changsha, Hunan 410004, China
| | - Guihua Wang
- Department of Oncology, Changsha Central Hospital, Changsha, Hunan 410004, China; Changsha Cancer Institute, Changsha Central Hospital, Changsha, Hunan 410004, China.
| |
Collapse
|
12
|
Muñoz-Rodríguez C, Fernández S, Osorio JM, Olivares F, Anfossi R, Bolivar S, Humeres C, Boza P, Vivar R, Pardo-Jimenez V, Hemmings KE, Turner NA, Díaz-Araya G. Expression and function of TLR4- induced B1R bradykinin receptor on cardiac fibroblasts. Toxicol Appl Pharmacol 2018; 351:46-56. [PMID: 29775649 DOI: 10.1016/j.taap.2018.05.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/05/2018] [Accepted: 05/10/2018] [Indexed: 12/22/2022]
Abstract
Cardiac fibroblasts (CF) are key cells for maintaining extracellular matrix (ECM) protein homeostasis in the heart, and for cardiac repair through CF-to-cardiac myofibroblast (CMF) differentiation. Additionally, CF play an important role in the inflammatory process after cardiac injury, and they express Toll like receptor 4 (TLR4), B1 and B2 bradykinin receptors (B1R and B2R) which are important in the inflammatory response. B1R and B2R are induced by proinflammatory cytokines and their activation by bradykinin (BK: B2R agonist) or des-arg-kallidin (DAKD: B1R agonist), induces NO and PGI2 production which is key for reducing collagen I levels. However, whether TLR4 activation regulates bradykinin receptor expression remains unknown. CF were isolated from human, neonatal rat and adult mouse heart. B1R mRNA expression was evaluated by qRT-PCR, whereas B1R, collagen, COX-2 and iNOS protein levels were evaluated by Western Blot. NO and PGI2 were evaluated by commercial kits. We report here that in CF, TLR4 activation increased B1R mRNA and protein levels, as well as COX-2 and iNOS levels. B1R mRNA levels were also induced by interleukin-1α via its cognate receptor IL-1R1. In LPS-pretreated CF the DAKD treatment induced higher responses with respect to those observed in non LPS-pretreated CF, increasing PGI2 secretion and NO production; and reducing collagen I protein levels in CF. In conclusion, no significant response to DAKD was observed (due to very low expression of B1R in CF) - but pre-activation of TLR4 in CF, conditions that significantly enhanced B1R expression, led to an additional response of DAKD.
Collapse
Affiliation(s)
- Claudia Muñoz-Rodríguez
- Laboratory of Molecular Pharmacology, Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences; University of Chile, Santiago, Chile
| | - Samuel Fernández
- Laboratory of Molecular Pharmacology, Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences; University of Chile, Santiago, Chile
| | - José Miguel Osorio
- Laboratory of Molecular Pharmacology, Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences; University of Chile, Santiago, Chile
| | - Francisco Olivares
- Laboratory of Molecular Pharmacology, Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences; University of Chile, Santiago, Chile
| | - Renatto Anfossi
- Laboratory of Molecular Pharmacology, Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences; University of Chile, Santiago, Chile
| | - Samir Bolivar
- Laboratory of Molecular Pharmacology, Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences; University of Chile, Santiago, Chile
| | - Claudio Humeres
- Laboratory of Molecular Pharmacology, Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences; University of Chile, Santiago, Chile
| | - Pía Boza
- Laboratory of Molecular Pharmacology, Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences; University of Chile, Santiago, Chile
| | - Raúl Vivar
- Laboratory of Molecular Pharmacology, Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences; University of Chile, Santiago, Chile
| | - Viviana Pardo-Jimenez
- Laboratory of Molecular Pharmacology, Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences; University of Chile, Santiago, Chile
| | - Karen E Hemmings
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Neil A Turner
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, Leeds LS2 9JT, United Kingdom.
| | - Guillermo Díaz-Araya
- Laboratory of Molecular Pharmacology, Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences; University of Chile, Santiago, Chile; Advanced Center of Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences; University of Chile, Santiago, Chile.
| |
Collapse
|