1
|
Thangaveloo M, Feng J, Phillips AR, Becker DL. Targeted Cx43 therapeutics reduce NLRP3 inflammasome activation in rat burn injury. Burns 2025; 51:107358. [PMID: 39798348 DOI: 10.1016/j.burns.2024.107358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 12/14/2024] [Accepted: 12/16/2024] [Indexed: 01/15/2025]
Abstract
Burns are dynamic injuries characterized by an initial zone of necrosis that progresses to compromise surrounding tissue. Acute inflammation and cell death are two main factors contributing to burn progression. These processes are modulated by Connexin43 (Cx43) hemichannels and gap junctions in burns and chronic wounds. Particularly, Cx43-mediated ATP release may interact with the P2X7 receptor to activate the NLRP3 inflammasome pathway. This study used a deep partial thickness rat burn model to evaluate the effect of Cx43 antisense oligodeoxynucleotides (Cx43asODN) or the Cx43 hemichannel blocker Tonabersat for the inhibition of inflammasome activation and their use as potential treatments for burn injury. Using immunofluorescence analysis, our data showed that Cx43asODN or Tonabersat reduced Cx43 hemichannel and gap junction expression. Concomitantly, they marginally and transiently reduced P2X7 expression and inflammasome complex assembly and inflammation. Quantitative analysis using H&E, Masson's trichrome & Picrosirus Red revealed reduced epidermal thickness and improved collagen preservation in treated burn wounds. Collectively, our findings suggest a possible involvement of the Cx43-mediated NLRP3 inflammasome pathway via P2X7 activation in early burn wound healing. This indicates that targeting Cx43 may have a potential therapeutic effect to improve healing outcomes.
Collapse
Affiliation(s)
- Moogaambikai Thangaveloo
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11, Mandalay Road, 308232, Singapore; Skin Research Institute Singapore, Level 17, Clinical Sciences Building, 11, Mandalay Road, 308232, Singapore
| | - Jiajun Feng
- Sengkang General Hospital, Singapore, Plastics, Reconstructive and Aesthetics Service, 110 Sengkang E Way, 544886, Singapore
| | - Anthony Rj Phillips
- School of Biological Sciences, The University of Auckland, 3A Symonds Street, Auckland 1010, New Zealand
| | - David L Becker
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11, Mandalay Road, 308232, Singapore; Skin Research Institute Singapore, Level 17, Clinical Sciences Building, 11, Mandalay Road, 308232, Singapore; National Skin Centre Singapore, 1 Mandalay Rd, 308205, Singapore.
| |
Collapse
|
2
|
Crowe LP, Gioseffi A, Bertolini MS, Docampo R. Inorganic Polyphosphate Is in the Surface of Trypanosoma cruzi but Is Not Significantly Secreted. Pathogens 2024; 13:776. [PMID: 39338967 PMCID: PMC11434814 DOI: 10.3390/pathogens13090776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 08/18/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Trypanosoma cruzi is the etiologic agent of Chagas disease, an infection that can lead to the development of cardiac fibrosis, which is characterized by the deposition of extracellular matrix (ECM) components in the interstitial region of the myocardium. The parasite itself can induce myofibroblast differentiation of cardiac fibroblast in vitro, leading to increased expression of ECM. Inorganic polyphosphate (polyP) is a linear polymer of orthophosphate that can also induce myofibroblast differentiation and deposition of ECM components and is highly abundant in T. cruzi. PolyP can modify proteins post-translationally by non-enzymatic polyphosphorylation of lysine residues of poly-acidic, serine-(S) and lysine (K)-rich (PASK) motifs. In this work, we used a bioinformatics screen and identified the presence of PASK domains in several surface proteins of T. cruzi. We also detected polyP in the external surface of its different life cycle stages and confirmed the stimulation of host cell fibrosis by trypomastigote infection. However, we were not able to detect significant secretion of the polymer or activation of transforming growth factor beta (TGF-β), an important factor for the generation of fibrosis by inorganic polyP- or trypomastigote-conditioned medium.
Collapse
Affiliation(s)
- Logan P. Crowe
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA (M.S.B.)
| | - Anna Gioseffi
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA (M.S.B.)
| | - Mayara S. Bertolini
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA (M.S.B.)
| | - Roberto Docampo
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA (M.S.B.)
- Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
3
|
Trovato F, Ceccarelli S, Michelini S, Vespasiani G, Guida S, Galadari HI, Nisticò SP, Colonna L, Pellacani G. Advancements in Regenerative Medicine for Aesthetic Dermatology: A Comprehensive Review and Future Trends. COSMETICS 2024; 11:49. [DOI: 10.3390/cosmetics11020049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
The growing interest in maintaining a youthful appearance has encouraged an accelerated development of innovative, minimally invasive aesthetic treatments for facial rejuvenation and regeneration. The close correlation between tissue repair, regeneration, and aging has paved the way for the application of regenerative medicine principles in cosmetic dermatology. The theoretical substrates of regenerative medicine applications in dermo-aesthetics are plentiful. However, regenerative dermatology is an emerging field and needs more data and in vivo trials to reach a consensus on the standardization of methods. In this review, we summarize the principles of regenerative medicine and techniques as they apply to cosmetic dermatology, suggesting unexplored fields and future directions.
Collapse
Affiliation(s)
- Federica Trovato
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Dermatology Clinic, Sapienza University of Rome, 00165 Rome, Italy
| | - Stefano Ceccarelli
- Department of Diagnostic and Laboratory Medicine, IRCCS Bambino Gesù Children’s Hospital, 00165 Rome, Italy
| | - Simone Michelini
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Dermatology Clinic, Sapienza University of Rome, 00165 Rome, Italy
| | - Giordano Vespasiani
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Dermatology Clinic, Sapienza University of Rome, 00165 Rome, Italy
| | - Stefania Guida
- Dermatology Department, Vita-Salute San Raffaele University, Via Olgettina n. 60, 20132 Milano, Italy
| | - Hassan Ibrahim Galadari
- College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
| | - Steven Paul Nisticò
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Dermatology Clinic, Sapienza University of Rome, 00165 Rome, Italy
| | - Laura Colonna
- Dermatology Unit, Istituto Dermopatico dell’Immacolata IDI-IRCCS, Via Monti di Creta 104, 00167 Rome, Italy
| | - Giovanni Pellacani
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Dermatology Clinic, Sapienza University of Rome, 00165 Rome, Italy
| |
Collapse
|
4
|
Tsuji Y, Ogata T, Mochizuki K, Tamura S, Morishita Y, Takamatsu T, Matoba S, Tanaka H. Myofibroblasts impair myocardial impulse propagation by heterocellular connexin43 gap-junctional coupling through micropores. Front Physiol 2024; 15:1352911. [PMID: 38465264 PMCID: PMC10920281 DOI: 10.3389/fphys.2024.1352911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 02/07/2024] [Indexed: 03/12/2024] Open
Abstract
Aim: Composite population of myofibroblasts (MFs) within myocardial tissue is known to alter impulse propagation, leading to arrhythmias. However, it remains unclear whether and how MFs alter their propagation patterns when contacting cardiomyocytes (CMs) without complex structural insertions in the myocardium. We attempted to unveil the effects of the one-sided, heterocellular CM-MF connection on the impulse propagation of CM monolayers without the spatial insertion of MFs as an electrical or mechanical obstacle. Methods and results: We evaluated fluo8-based spatiotemporal patterns in impulse propagation of neonatal rat CM monolayers cultured on the microporous membrane having 8-μm diameter pores with co-culture of MFs or CMs on the reverse membrane side (CM-MF model or CM-CM model, respectively). During consecutive pacing at 1 or 2 Hz, the CM monolayers exhibited forward impulse propagation from the pacing site with a slower conduction velocity (θ) and a larger coefficient of directional θ variation in the CM-MF model than that in the CM-CM model in a frequency-dependent manner (2 Hz >1 Hz). The localized placement of an MF cluster on the reverse side resulted in an abrupt segmental depression of the impulse propagation of the upper CM layer, causing a spatiotemporally non-uniform pattern. Dye transfer of the calcein loaded in the upper CM layer to the lower MF layer was attenuated by the gap-junction inhibitor heptanol. Immunocytochemistry identified definitive connexin 43 (Cx43) between the CMs and MFs in the membrane pores. MF-selective Cx43 knockdown in the MF layer improved both the velocity and uniformity of propagation in the CM monolayer. Conclusion: Heterocellular Cx43 gap junction coupling of CMs with MFs alters the spatiotemporal patterns of myocardial impulse propagation, even in the absence of spatially interjacent and mechanosensitive modulations by MFs. Moreover, MFs can promote pro-arrhythmogenic impulse propagation when in face-to-face contact with the myocardium that arises in the healing infarct border zone.
Collapse
Affiliation(s)
- Yumika Tsuji
- Department of Pathology and Cell Regulation and, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takehiro Ogata
- Department of Pathology and Cell Regulation and, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kentaro Mochizuki
- Department of Pathology and Cell Regulation and, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shoko Tamura
- Department of Pathology and Cell Regulation and, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuma Morishita
- Department of Pathology and Cell Regulation and, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tetsuro Takamatsu
- Department of Pathology and Cell Regulation and, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Medical Photonics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hideo Tanaka
- Department of Pathology and Cell Regulation and, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Faculty of Health and Medical Sciences, Kyoto University of Advanced Science, Kyoto, Japan
| |
Collapse
|
5
|
Pavic B, Ogorevc M, Boric K, Vukovic D, Saraga-Babic M, Mardesic S. Connexin 37, 40, 43 and Pannexin 1 Expression in the Gastric Mucosa of Patients with Systemic Sclerosis. Biomedicines 2023; 11:2487. [PMID: 37760928 PMCID: PMC10525958 DOI: 10.3390/biomedicines11092487] [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: 08/09/2023] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disease characterized by fibrosis of the skin and internal organs. Although its pathogenesis is not fully understood, connexins (Cxs) and pannexins (Panx) could be involved in the process of fibrosis. We analyzed the protein expression of Cx37, Cx40, Cx43, and Panx1 in the gastric mucosa of patients with SSc and healthy volunteers, using immunofluorescence staining. Protein levels of Cx37 were slightly increased, while the levels of Cx40 were significantly decreased in the lamina propria of the gastric mucosa of SSc patients compared to the controls. The changes were proportional to SSc severity, with the most prominent changes found in patients with severe diffuse cutaneous SSc. No differences in Cx43 or Panx1 levels were found between the analyzed groups of samples. The lack of changes in Cx43 expression, which has been previously associated with fibrosis, could be due to the weak expression of Cx43 in the gastric mucosa in general. Further studies on full-thickness gastric biopsies containing muscle layers and animal SSc models are needed to fully elucidate the role of Cxs and Panxs in SSc-associated fibrosis.
Collapse
Affiliation(s)
- Berna Pavic
- Renal Unit, University Hospital of Split, Šoltanska 1, 21000 Split, Croatia;
| | - Marin Ogorevc
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia; (M.O.); (M.S.-B.)
| | - Katarina Boric
- Department of Internal Medicine, University Hospital of Split, Šoltanska 1, 21000 Split, Croatia;
| | - Dubravka Vukovic
- Department of Dermatovenerology, University Hospital of Split, Šoltanska 1, 21000 Split, Croatia;
| | - Mirna Saraga-Babic
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia; (M.O.); (M.S.-B.)
| | - Snjezana Mardesic
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21000 Split, Croatia; (M.O.); (M.S.-B.)
| |
Collapse
|
6
|
Zhou E, Zhou J, Bi C, Zhang Z. Cx43 Facilitates Mesenchymal Transition of Endothelial Cells Induced by Shear Stress. J Vasc Res 2023; 60:204-212. [PMID: 37673049 PMCID: PMC10614473 DOI: 10.1159/000533320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/28/2023] [Indexed: 09/08/2023] Open
Abstract
OBJECTIVES This study aimed to determine the function of Cx43 in the endothelial-to-mesenchymal transition (EndMT) process of endothelial cells (ECs) and to explore the potential signaling pathways underlying these functions. METHODS ECs were extracted from rat aorta. ECs were transfected with Cx43 cDNA and Cx43 siRNA and then exposed to 5 or 12 dyne/cm2. Immunofluorescence staining was used to detect the expression of SM22α, Cx43, and acetylated α-tubulin in ECs. Western blotting was used to detect the protein expression of α-SMA, CD31, Cx43, H1-calponin, Ift88, and p-smad3 in ECs. RESULTS The expression of αSMA, SM22α, and Cx43 was significantly increased, and CD31 was markedly decreased in ECs treated with laminar shear stress at 5 dyn/cm2. The Cx43 cDNA transfection could induce the expression of SM22α or H1-calponin and attenuate CD31 expression in ECs. Also, Cx43 overexpression harms cilia formation in ECs exposed to 5 dyn/cm2, accompanied with the regulated Ift88 and smad signaling. CONCLUSIONS This study found that laminar shear stress at 5 dyn/cm2 would increase the expression of Cx43 to facilitate the EndMT process of ECs, associated with morphological changes in primary cilia and the decreased expression of Ift88 in ECs.
Collapse
Affiliation(s)
- En Zhou
- Department of Cardiovascular Surgery, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Zhou
- Department of Cardiovascular Surgery, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Changlong Bi
- Department of Cardiology, Central Hospital of Minhang District, Shanghai, China
| | - Zongqi Zhang
- Department of Cardiovascular Surgery, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
7
|
Chua JW, Thangaveloo M, Lim DXE, Madden LE, Phillips ARJ, Becker DL. Connexin43 in Post-Surgical Peritoneal Adhesion Formation. Life (Basel) 2022; 12:1734. [PMID: 36362888 PMCID: PMC9697983 DOI: 10.3390/life12111734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/15/2022] [Accepted: 10/26/2022] [Indexed: 09/10/2024] Open
Abstract
OBJECTIVE Post-surgical peritoneal adhesions are a serious problem for the quality of life and fertility. Yet there are no effective ways of preventing their occurrence. The gap junction protein Cx43 is known to be involved in fibrosis in several different organs and disease conditions often associated with inflammation. Here we examined the Cx43 dynamic expression in an ischemic button model of surgical adhesions. METHODS Using the mouse ischemic button model, Cx43 antisense was delivered in Pluronic gel to attenuate Cx43 expression. The severity of button formation and immunofluorescence analysis of Cx43 and TGF-β1 were performed. The concentration of tissue plasminogen activator via ELISA was also performed. RESULTS As early as 6 h after button formation, the Cx43 levels were elevated in and around the button and some weak adhesions were formed. By 24 h Cx43 levels had increased further and adhesions were more defined. At 7 days the adhesions were much more robust, opaque, and vascularized, requiring blunt or sharp dissection to break them. Cx43 antisense attenuated its upregulation and, reduced the number and severity of adhesions that formed. CONCLUSION Targeting Cx43 after surgical procedures may be a potential therapeutic strategy for preventing adhesion formation or at least reducing their severity.
Collapse
Affiliation(s)
- Jia Wang Chua
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232, Singapore
- Skin Research Institute Singapore, Singapore 308232, Singapore
| | - Moogaambikai Thangaveloo
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232, Singapore
- Skin Research Institute Singapore, Singapore 308232, Singapore
| | - Debbie Xiu En Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232, Singapore
- Skin Research Institute Singapore, Singapore 308232, Singapore
| | - Leigh E. Madden
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232, Singapore
- Skin Research Institute Singapore, Singapore 308232, Singapore
| | | | - David L. Becker
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore 308232, Singapore
- Skin Research Institute Singapore, Singapore 308232, Singapore
| |
Collapse
|
8
|
Shi Y, Li X, Yang J. Cx43 upregulation in HUVECs under stretch via TGF-β1 and cytoskeletal network. Open Med (Wars) 2022; 17:463-474. [PMID: 35350835 PMCID: PMC8919824 DOI: 10.1515/med-2022-0432] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/25/2022] Open
Abstract
Many physiological and pathophysiological processes in cells or tissues are involved in mechanical stretch, which induces the gap junction gene expression and cytokine TGF beta changes. However, the underlying mechanisms of the gap junction gene expression remain unknown. Here, we showed that the mRNA and protein levels of Cx43 in human umbilical vein endothelial cells (HUVECs) were significantly increased after 24 h stretch stimulation, and TGF beta1 (not TGF beta2) expression was also upregulated. Administration of TGF beta1 into HUVECs without stretch also induced upregulation of Cx43 expression. However, SB431542, a specific inhibitor of the TGF beta1 receptor, blocked the Cx43 protein upregulation caused by TGF beta1. Further, the increase of Cx43 protein expression under the stretch condition was partially blocked by SB431542; it was also partially blocked by simultaneous administration of anti-TGF beta1 monoclonal neutralization antibody. Importantly, the upregulation of Cx43 induced by stretch was blocked by the administration of actin and microtubule inhibitors, while NEDD4, a key element in mediating Cx43 protein ubiquitination and degradation, was not changed under the stretch condition. In conclusion, upregulation of Cx43 expression under the 24 h stretch condition is mediated via TGF beta1 receptor signaling pathway, and it also involves the actin and microtubule cytoskeletal network.
Collapse
Affiliation(s)
- Yumeng Shi
- Department of Ophthalmology and Visual Science, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai 200031, China
| | - Xinbo Li
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Oregon, USA
| | - Jin Yang
- Department of Ophthalmology and Visual Science, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai 200031, China
| |
Collapse
|
9
|
Du X. Sympatho-adrenergic mechanisms in heart failure: new insights into pathophysiology. MEDICAL REVIEW (BERLIN, GERMANY) 2021; 1:47-77. [PMID: 37724075 PMCID: PMC10388789 DOI: 10.1515/mr-2021-0007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/02/2021] [Indexed: 09/20/2023]
Abstract
The sympathetic nervous system is activated in the setting of heart failure (HF) to compensate for hemodynamic instability. However, acute sympathetic surge or sustained high neuronal firing rates activates β-adrenergic receptor (βAR) signaling contributing to myocardial remodeling, dysfunction and electrical instability. Thus, sympatho-βAR activation is regarded as a hallmark of HF and forms pathophysiological basis for β-blocking therapy. Building upon earlier research findings, studies conducted in the recent decades have significantly advanced our understanding on the sympatho-adrenergic mechanism in HF, which forms the focus of this article. This review notes recent research progress regarding the roles of cardiac β2AR or α1AR in the failing heart, significance of β1AR-autoantibodies, and βAR signaling through G-protein independent signaling pathways. Sympatho-βAR regulation of immune cells or fibroblasts is specifically discussed. On the neuronal aspects, knowledge is assembled on the remodeling of sympathetic nerves of the failing heart, regulation by presynaptic α2AR of NE release, and findings on device-based neuromodulation of the sympathetic nervous system. The review ends with highlighting areas where significant knowledge gaps exist but hold promise for new breakthroughs.
Collapse
Affiliation(s)
- Xiaojun Du
- Faculty of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, 76 West Yanta Road, Xi’an710061, Shaanxi, China
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC3004, Australia
| |
Collapse
|
10
|
Martins-Marques T. Connecting different heart diseases through intercellular communication. Biol Open 2021; 10:bio058777. [PMID: 34494646 PMCID: PMC8443862 DOI: 10.1242/bio.058777] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 07/12/2021] [Indexed: 12/22/2022] Open
Abstract
Well-orchestrated intercellular communication networks are pivotal to maintaining cardiac homeostasis and to ensuring adaptative responses and repair after injury. Intracardiac communication is sustained by cell-cell crosstalk, directly via gap junctions (GJ) and tunneling nanotubes (TNT), indirectly through the exchange of soluble factors and extracellular vesicles (EV), and by cell-extracellular matrix (ECM) interactions. GJ-mediated communication between cardiomyocytes and with other cardiac cell types enables electrical impulse propagation, required to sustain synchronized heart beating. In addition, TNT-mediated organelle transfer has been associated with cardioprotection, whilst communication via EV plays diverse pathophysiological roles, being implicated in angiogenesis, inflammation and fibrosis. Connecting various cell populations, the ECM plays important functions not only in maintaining the heart structure, but also acting as a signal transducer for intercellular crosstalk. Although with distinct etiologies and clinical manifestations, intercellular communication derailment has been implicated in several cardiac disorders, including myocardial infarction and hypertrophy, highlighting the importance of a comprehensive and integrated view of complex cell communication networks. In this review, I intend to provide a critical perspective about the main mechanisms contributing to regulate cellular crosstalk in the heart, which may be considered in the development of future therapeutic strategies, using cell-based therapies as a paradigmatic example. This Review has an associated Future Leader to Watch interview with the author.
Collapse
Affiliation(s)
- Tania Martins-Marques
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, 3000-548 Coimbra, Portugal
- Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Centre of Coimbra (CACC), 3004-561 Coimbra, Portugal
| |
Collapse
|
11
|
Kostecki GM, Shi Y, Chen CS, Reich DH, Entcheva E, Tung L. Optogenetic current in myofibroblasts acutely alters electrophysiology and conduction of co-cultured cardiomyocytes. Sci Rep 2021; 11:4430. [PMID: 33627695 PMCID: PMC7904933 DOI: 10.1038/s41598-021-83398-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/27/2021] [Indexed: 01/31/2023] Open
Abstract
Interactions between cardiac myofibroblasts and myocytes may slow conduction and generate spontaneous beating in fibrosis, increasing the chance of life-threatening arrhythmia. While co-culture studies have shown that myofibroblasts can affect cardiomyocyte electrophysiology in vitro, the extent of myofibroblast-myocyte electrical conductance in a syncytium is unknown. In this neonatal rat study, cardiac myofibroblasts were transduced with Channelrhodopsin-2, which allowed acute and selective increase of myofibroblast current, and plated on top of cardiomyocytes. Optical mapping revealed significantly decreased conduction velocity (- 27 ± 6%, p < 10-3), upstroke rate (- 13 ± 4%, p = 0.002), and action potential duration (- 14 ± 7%, p = 0.004) in co-cultures when 0.017 mW/mm2 light was applied, as well as focal spontaneous beating in 6/7 samples and a decreased cycle length (- 36 ± 18%, p = 0.002) at 0.057 mW/mm2 light. In silico modeling of the experiments reproduced the experimental findings and suggested the light levels used in experiments produced excess current similar in magnitude to endogenous myofibroblast current. Fitting the model to experimental data predicted a tissue-level electrical conductance across the 3-D interface between myofibroblasts and cardiomyocytes of ~ 5 nS/cardiomyocyte, and showed how increased myofibroblast-myocyte conductance, increased myofibroblast/myocyte capacitance ratio, and increased myofibroblast current, which occur in fibrosis, can work in tandem to produce pro-arrhythmic increases in conduction and spontaneous beating.
Collapse
Affiliation(s)
- Geran M Kostecki
- Department of Biomedical Engineering, Johns Hopkins University, 720 Rutland Ave., Baltimore, MD, 21205, USA
| | - Yu Shi
- Department of Physics and Astronomy, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Christopher S Chen
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Daniel H Reich
- Department of Physics and Astronomy, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Washington, DC, USA
| | - Leslie Tung
- Department of Biomedical Engineering, Johns Hopkins University, 720 Rutland Ave., Baltimore, MD, 21205, USA.
| |
Collapse
|
12
|
Zhao Y, Iyer S, Tavanaei M, Nguyen NT, Lin A, Nguyen TP. Proarrhythmic Electrical Remodeling by Noncardiomyocytes at Interfaces With Cardiomyocytes Under Oxidative Stress. Front Physiol 2021; 11:622613. [PMID: 33603677 PMCID: PMC7884825 DOI: 10.3389/fphys.2020.622613] [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: 10/29/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022] Open
Abstract
Life-threatening ventricular arrhythmias, typically arising from interfaces between fibrosis and surviving cardiomyocytes, are feared sequelae of structurally remodeled hearts under oxidative stress. Incomplete understanding of the proarrhythmic electrical remodeling by fibrosis limits the development of novel antiarrhythmic strategies. To define the mechanistic determinants of the proarrhythmia in electrical crosstalk between cardiomyocytes and noncardiomyocytes, we developed a novel in vitro model of interface between neonatal rat ventricular cardiomyocytes (NRVMs) and controls [NRVMs or connexin43 (Cx43)-deficient HeLa cells] vs. Cx43+ noncardiomyocytes [aged rat ventricular myofibroblasts (ARVFs) or HeLaCx43 cells]. We performed high-speed voltage-sensitive optical imaging at baseline and following acute H2O2 exposure. In NRVM-NRVM and NRVM-HeLa controls, no arrhythmias occurred under either experimental condition. In the NRVM-ARVF and NRVM-HeLaCx43 groups, Cx43+ noncardiomyocytes enabled passive decremental propagation of electrical impulses and impaired NRVM activation and repolarization, thereby slowing conduction and prolonging action potential duration. Following H2O2 exposure, arrhythmia triggers, automaticity, and non-reentrant and reentrant arrhythmias emerged. This study reveals that myofibroblasts (which generate cardiac fibrosis) and other noncardiomyocytes can induce not only structural remodeling but also electrical remodeling and that electrical remodeling by noncardiomyocytes can be particularly arrhythmogenic in the presence of an oxidative burst. Synergistic electrical remodeling between H2O2 and noncardiomyocytes may account for the clinical arrhythmogenicity of myofibroblasts at fibrotic interfaces with cardiomyocytes in ischemic/non-ischemic cardiomyopathies. Understanding the enhanced arrhythmogenicity of synergistic electrical remodeling by H2O2 and noncardiomyocytes may guide novel safe-by-design antiarrhythmic strategies for next-generation iatrogenic interfaces between surviving native cardiomyocytes and exogenous stem cells or engineered tissues in cardiac regenerative therapies.
Collapse
Affiliation(s)
- Yali Zhao
- Division of Cardiology, Department of Medicine, The Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Shankar Iyer
- Division of Cardiology, Department of Medicine, The Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Maryam Tavanaei
- Division of Cardiology, Department of Medicine, The Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Nicole T Nguyen
- Division of Cardiology, Department of Medicine, The Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Andrew Lin
- Division of Cardiology, Department of Medicine, The Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Thao P Nguyen
- Division of Cardiology, Department of Medicine, The Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| |
Collapse
|
13
|
Human Cardiac Fibroblast Number and Activation State Modulate Electromechanical Function of hiPSC-Cardiomyocytes in Engineered Myocardium. Stem Cells Int 2020; 2020:9363809. [PMID: 32724316 PMCID: PMC7381987 DOI: 10.1155/2020/9363809] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 05/11/2020] [Indexed: 11/27/2022] Open
Abstract
Cardiac tissue engineering using hiPSC-derived cardiomyocytes is a promising avenue for cardiovascular regeneration, pharmaceutical drug development, cardiotoxicity evaluation, and disease modeling. Limitations to these applications still exist due in part to the need for more robust structural support, organization, and electromechanical function of engineered cardiac tissues. It is well accepted that heterotypic cellular interactions impact the phenotype of cardiomyocytes. The current study evaluates the functional effects of coculturing adult human cardiac fibroblasts (hCFs) in 3D engineered tissues on excitation and contraction with the goal of recapitulating healthy, nonarrhythmogenic myocardium in vitro. A small population (5% of total cell number) of hCFs in tissues improves tissue formation, material properties, and contractile function. However, two perturbations to the hCF population create disease-like phenotypes in engineered cardiac tissues. First, increasing the percentage of hCFs to 15% resulted in tissues with increased ectopic activity and spontaneous excitation rate. Second, hCFs undergo myofibroblast activation in traditional two-dimensional culture, and this altered phenotype ablated the functional benefits of hCFs when incorporated into engineered cardiac tissues. Taken together, the results of this study demonstrate that human cardiac fibroblast number and activation state modulate electromechanical function of hiPSC-cardiomyocytes and that a low percentage of quiescent hCFs are a valuable cell source to advance a healthy electromechanical response of engineered cardiac tissue for regenerative medicine applications.
Collapse
|
14
|
Squecco R, Chellini F, Idrizaj E, Tani A, Garella R, Pancani S, Pavan P, Bambi F, Zecchi-Orlandini S, Sassoli C. Platelet-Rich Plasma Modulates Gap Junction Functionality and Connexin 43 and 26 Expression During TGF-β1-Induced Fibroblast to Myofibroblast Transition: Clues for Counteracting Fibrosis. Cells 2020; 9:cells9051199. [PMID: 32408529 PMCID: PMC7290305 DOI: 10.3390/cells9051199] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/28/2020] [Accepted: 05/08/2020] [Indexed: 12/19/2022] Open
Abstract
Skeletal muscle repair/regeneration may benefit by Platelet-Rich Plasma (PRP) treatment owing to PRP pro-myogenic and anti-fibrotic effects. However, PRP anti-fibrotic action remains controversial. Here, we extended our previous researches on the inhibitory effects of PRP on in vitro transforming growth factor (TGF)-β1-induced differentiation of fibroblasts into myofibroblasts, the effector cells of fibrosis, focusing on gap junction (GJ) intercellular communication. The myofibroblastic phenotype was evaluated by cell shape analysis, confocal fluorescence microscopy and Western blotting analyses of α-smooth muscle actin and type-1 collagen expression, and electrophysiological recordings of resting membrane potential, resistance, and capacitance. PRP negatively regulated myofibroblast differentiation by modifying all the assessed parameters. Notably, myofibroblast pairs showed an increase of voltage-dependent GJ functionality paralleled by connexin (Cx) 43 expression increase. TGF-β1-treated cells, when exposed to a GJ blocker, or silenced for Cx43 expression, failed to differentiate towards myofibroblasts. Although a minority, myofibroblast pairs also showed not-voltage-dependent GJ currents and coherently Cx26 expression. PRP abolished the TGF-β1-induced voltage-dependent GJ current appearance while preventing Cx43 increase and promoting Cx26 expression. This study adds insights into molecular and functional mechanisms regulating fibroblast-myofibroblast transition and supports the anti-fibrotic potential of PRP, demonstrating the ability of this product to hamper myofibroblast generation targeting GJs.
Collapse
Affiliation(s)
- Roberta Squecco
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy; (R.S.); (E.I.); (R.G.)
| | - Flaminia Chellini
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (S.P.); (S.Z.-O.)
| | - Eglantina Idrizaj
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy; (R.S.); (E.I.); (R.G.)
| | - Alessia Tani
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (S.P.); (S.Z.-O.)
| | - Rachele Garella
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy; (R.S.); (E.I.); (R.G.)
| | - Sofia Pancani
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (S.P.); (S.Z.-O.)
| | - Paola Pavan
- Transfusion Medicine and Cell Therapy Unit, "A. Meyer" University Children’s Hospital, 50134 Florence, Italy; (P.P.); (F.B.)
| | - Franco Bambi
- Transfusion Medicine and Cell Therapy Unit, "A. Meyer" University Children’s Hospital, 50134 Florence, Italy; (P.P.); (F.B.)
| | - Sandra Zecchi-Orlandini
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (S.P.); (S.Z.-O.)
| | - Chiara Sassoli
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (S.P.); (S.Z.-O.)
- Correspondence: ; Tel.: +39-0552-7580-63
| |
Collapse
|
15
|
Le LV, Mkrtschjan MA, Russell B, Desai TA. Hang on tight: reprogramming the cell with microstructural cues. Biomed Microdevices 2019; 21:43. [PMID: 30955102 PMCID: PMC6791714 DOI: 10.1007/s10544-019-0394-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cells interact intimately with complex microdomains in their extracellular matrix (ECM) and maintain a delicate balance of mechanical forces through mechanosensitive cellular components. Tissue injury results in acute degradation of the ECM and disruption of cell-ECM contacts, manifesting in loss of cytoskeletal tension, leading to pathological cell transformation and the onset of disease. Recently, microscale hydrogel constructs have been developed to provide cells with microdomains to form focal adhesion binding sites, which enable restoration of cytoskeletal tension. These synthetic anchors can recapitulate the complex 3D architecture of the native ECM to provide microtopographical cues. The mechanical deformation of proteins at the cell surface can activate signaling cascades to modulate downstream gene-level transcription, making this a unique materials-based approach for reprogramming cell behavior. An overview of the mechanisms underlying these mechanosensitive interactions in fibroblasts, stem and other cell types is provided to review their effects on cellular reprogramming. Recent investigations on the fabrication, functionalization and implementation of these materials and microtopographical features for drug testing and therapeutic applications are discussed.
Collapse
Affiliation(s)
- Long V Le
- Department of Bioengineering and Therapeutic Sciences, University of California, 1700 4th St Rm 204, San Francisco, CA, 94158, USA
| | - Michael A Mkrtschjan
- Department of Bioengineering, University of Illinois, Chicago, 835 S. Wolcott, Chicago, IL, 60612, USA
| | - Brenda Russell
- Department of Physiology and Biophysics, University of Illinois, Chicago, 835 S. Wolcott, Chicago, IL, 60612, USA
| | - Tejal A Desai
- Department of Bioengineering and Therapeutic Sciences, University of California, 1700 4th St Rm 204, San Francisco, CA, 94158, USA.
| |
Collapse
|
16
|
Chen Q, Gimple RC, Li G, Chen J, Wu H, Li R, Xie J, Xu B. LIM kinase 1 acts as a profibrotic mediator in permanent atrial fibrillation patients with valvular heart disease. J Biosci 2019. [DOI: 10.1007/s12038-018-9825-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
17
|
Michalik M, Wójcik-Pszczoła K, Paw M, Wnuk D, Koczurkiewicz P, Sanak M, Pękala E, Madeja Z. Fibroblast-to-myofibroblast transition in bronchial asthma. Cell Mol Life Sci 2018; 75:3943-3961. [PMID: 30101406 PMCID: PMC6182337 DOI: 10.1007/s00018-018-2899-4] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 07/26/2018] [Accepted: 08/06/2018] [Indexed: 12/11/2022]
Abstract
Bronchial asthma is a chronic inflammatory disease in which bronchial wall remodelling plays a significant role. This phenomenon is related to enhanced proliferation of airway smooth muscle cells, elevated extracellular matrix protein secretion and an increased number of myofibroblasts. Phenotypic fibroblast-to-myofibroblast transition represents one of the primary mechanisms by which myofibroblasts arise in fibrotic lung tissue. Fibroblast-to-myofibroblast transition requires a combination of several types of factors, the most important of which are divided into humoural and mechanical factors, as well as certain extracellular matrix proteins. Despite intensive research on the nature of this process, its underlying mechanisms during bronchial airway wall remodelling in asthma are not yet fully clarified. This review focuses on what is known about the nature of fibroblast-to-myofibroblast transition in asthma. We aim to consider possible mechanisms and conditions that may play an important role in fibroblast-to-myofibroblast transition but have not yet been discussed in this context. Recent studies have shown that some inherent and previously undescribed features of fibroblasts can also play a significant role in fibroblast-to-myofibroblast transition. Differences observed between asthmatic and non-asthmatic bronchial fibroblasts (e.g., response to transforming growth factor β, cell shape, elasticity, and protein expression profile) may have a crucial influence on this phenomenon. An accurate understanding and recognition of all factors affecting fibroblast-to-myofibroblast transition might provide an opportunity to discover efficient methods of counteracting this phenomenon.
Collapse
Affiliation(s)
- Marta Michalik
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| | - Katarzyna Wójcik-Pszczoła
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland.
| | - Milena Paw
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Dawid Wnuk
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Paulina Koczurkiewicz
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Marek Sanak
- Division of Molecular Biology and Clinical Genetics, Department of Medicine, Jagiellonian University Medical College, Skawińska 8, 31-066, Kraków, Poland
| | - Elżbieta Pękala
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Zbigniew Madeja
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| |
Collapse
|
18
|
Chellini F, Tani A, Vallone L, Nosi D, Pavan P, Bambi F, Zecchi Orlandini S, Sassoli C. Platelet-Rich Plasma Prevents In Vitro Transforming Growth Factor-β1-Induced Fibroblast to Myofibroblast Transition: Involvement of Vascular Endothelial Growth Factor (VEGF)-A/VEGF Receptor-1-Mediated Signaling †. Cells 2018; 7:cells7090142. [PMID: 30235859 PMCID: PMC6162453 DOI: 10.3390/cells7090142] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/14/2018] [Accepted: 09/16/2018] [Indexed: 02/07/2023] Open
Abstract
The antifibrotic potential of platelet-rich plasma (PRP) is controversial. This study examined the effects of PRP on in vitro transforming growth factor (TGF)-β1-induced differentiation of fibroblasts into myofibroblasts, the main drivers of fibrosis, and the involvement of vascular endothelial growth factor (VEGF)-A in mediating PRP-induced responses. The impact of PRP alone on fibroblast differentiation was also assessed. Myofibroblastic phenotype was evaluated by confocal fluorescence microscopy and western blotting analyses of α-smooth muscle actin (sma) and type-1 collagen expression, vinculin-rich focal adhesion clustering, and stress fiber assembly. Notch-1, connexin 43, and VEGF-A expression were also analyzed by RT-PCR. PRP negatively regulated fibroblast-myofibroblast transition via VEGF-A/VEGF receptor (VEGFR)-1-mediated inhibition of TGF-β1/Smad3 signaling. Indeed TGF-β1/PRP co-treated fibroblasts showed a robust attenuation of the myofibroblastic phenotype concomitant with a decrease of Smad3 expression levels. The VEGFR-1 inhibition by KRN633 or blocking antibodies, or VEGF-A neutralization in these cells prevented the PRP-promoted effects. Moreover PRP abrogated the TGF-β1-induced reduction of VEGF-A and VEGFR-1 cell expression. The role of VEGF-A signaling in counteracting myofibroblast generation was confirmed by cell treatment with soluble VEGF-A. PRP as single treatment did not induce fibroblast myodifferentiation. This study provides new insights into cellular and molecular mechanisms underpinning PRP antifibrotic action.
Collapse
Affiliation(s)
- Flaminia Chellini
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy.
| | - Alessia Tani
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy.
| | - Larissa Vallone
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy.
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy.
| | - Paola Pavan
- Transfusion Medicine and Cell Therapy Unit, "A. Meyer" University Children's Hospital, 50139 Florence, Italy.
| | - Franco Bambi
- Transfusion Medicine and Cell Therapy Unit, "A. Meyer" University Children's Hospital, 50139 Florence, Italy.
| | - Sandra Zecchi Orlandini
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy.
| | - Chiara Sassoli
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy.
| |
Collapse
|
19
|
Cao L, Chen Y, Lu L, Liu Y, Wang Y, Fan J, Yin Y. Angiotensin II upregulates fibroblast-myofibroblast transition through Cx43-dependent CaMKII and TGF-β1 signaling in neonatal rat cardiac fibroblasts. Acta Biochim Biophys Sin (Shanghai) 2018; 50:843-852. [PMID: 30060053 DOI: 10.1093/abbs/gmy090] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/05/2018] [Indexed: 02/07/2023] Open
Abstract
In cardiac fibroblasts, angiotensin II (Ang II) can increase connexin 43 (Cx43) expression and promote calmodulin-dependent protein kinase II (CaMKII) activation. Cx43 overexpression is crucial for the fibroblast-myofibroblast transition. The main purpose of the present study was to investigate the role of CaMKII in regulating Cx43 expression and to determine whether the CaMKII/Cx43 pathway is essential for controlling fibroblast activation and differentiation. In vivo, 4 weeks of Ang II infusion enhanced CaMKII activation but reduced Cx43 expression in hearts undergoing fibrosis remodeling, while in cultured neonatal rat fibroblasts, CaMKII activation upregulated Cx43 expression via transforming growth factor-beta1 (TGF-β1). CaMKII inhibition by Ang-(1-7) or autocamtide 2-related inhibitory peptide reversed the Ang II-induced changes in Cx43 expression and attenuated Ang II-induced upregulation of alpha smooth muscle actin and TGF-β1 in both Ang II-infused rats and cultured fibroblasts. Based on the in vivo and in vitro experimental results, CaMKII plays a pivotal role in the Ang II-mediated fibroblast-myofibroblast transition by modulating the expressions of TGF-β1 and Cx43. We conclude that Ang II mediates the fibroblast-myofibroblast transition partially via the Ang II/CaMKII/TGF-β1/Cx43 signaling pathway.
Collapse
Affiliation(s)
- Li Cao
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunlin Chen
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Lu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yihao Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yaowen Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinqi Fan
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuehui Yin
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
20
|
Paw M, Wnuk D, Kądziołka D, Sęk A, Lasota S, Czyż J, Madeja Z, Michalik M. Fenofibrate Reduces the Asthma-Related Fibroblast-To-Myofibroblast Transition by TGF-Β/Smad2/3 Signaling Attenuation and Connexin 43-Dependent Phenotype Destabilization. Int J Mol Sci 2018; 19:ijms19092571. [PMID: 30158495 PMCID: PMC6163263 DOI: 10.3390/ijms19092571] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 08/21/2018] [Accepted: 08/27/2018] [Indexed: 12/26/2022] Open
Abstract
The activation of human bronchial fibroblasts by transforming growth factor-β1 (TGF-β1) leads to the formation of highly contractile myofibroblasts in the process of the fibroblast–myofibroblast transition (FMT). This process is crucial for subepithelial fibrosis and bronchial wall remodeling in asthma. However, this process evades current therapeutic asthma treatment strategies. Since our previous studies showed the attenuation of the TGF-β1-induced FMT in response to lipid-lowering agents (e.g., statins), we were interested to see whether a corresponding effect could be obtained upon administration of hypolipidemic agents. In this study, we investigated the effect of fenofibrate on FMT efficiency in populations of bronchial fibroblasts derived from asthmatic patients. Fenofibrate exerted a dose-dependent inhibitory effect on the FMT, even though it did not efficiently affect the expression of α-smooth muscle actin (α-SMA; marker of myofibroblasts); however, it considerably reduced its incorporation into stress fibers through connexin 43 regulation. This effect was accompanied by disturbances in the actin cytoskeleton architecture, impairments in the maturation of focal adhesions, and the fenofibrate-induced deactivation of TGF-β1/Smad2/3 signaling. These data suggest that fenofibrate interferes with myofibroblastic differentiation during asthma-related subepithelial fibrosis. The data indicate the potential application of fenofibrate in the therapy and prevention of bronchial remodeling during the asthmatic process.
Collapse
Affiliation(s)
- Milena Paw
- Faculty of Biophysics, Biochemistry and Biotechnology, Department of Cell Biology, Jagiellonian University, Gronostajowa 7, 30-378 Kraków, Poland.
| | - Dawid Wnuk
- Faculty of Biophysics, Biochemistry and Biotechnology, Department of Cell Biology, Jagiellonian University, Gronostajowa 7, 30-378 Kraków, Poland.
| | - Dominika Kądziołka
- Faculty of Biophysics, Biochemistry and Biotechnology, Department of Cell Biology, Jagiellonian University, Gronostajowa 7, 30-378 Kraków, Poland.
| | - Aleksandra Sęk
- Faculty of Biophysics, Biochemistry and Biotechnology, Department of Cell Biology, Jagiellonian University, Gronostajowa 7, 30-378 Kraków, Poland.
- Nencki Institute of Experimental Biology, Laboratory of Intracellular Ion Channels, 02-093 Warsaw, Poland.
| | - Sławomir Lasota
- Faculty of Biophysics, Biochemistry and Biotechnology, Department of Cell Biology, Jagiellonian University, Gronostajowa 7, 30-378 Kraków, Poland.
| | - Jarosław Czyż
- Faculty of Biophysics, Biochemistry and Biotechnology, Department of Cell Biology, Jagiellonian University, Gronostajowa 7, 30-378 Kraków, Poland.
| | - Zbigniew Madeja
- Faculty of Biophysics, Biochemistry and Biotechnology, Department of Cell Biology, Jagiellonian University, Gronostajowa 7, 30-378 Kraków, Poland.
| | - Marta Michalik
- Faculty of Biophysics, Biochemistry and Biotechnology, Department of Cell Biology, Jagiellonian University, Gronostajowa 7, 30-378 Kraków, Poland.
| |
Collapse
|
21
|
March JT, Golshirazi G, Cernisova V, Carr H, Leong Y, Lu-Nguyen N, Popplewell LJ. Targeting TGFβ Signaling to Address Fibrosis Using Antisense Oligonucleotides. Biomedicines 2018; 6:biomedicines6030074. [PMID: 29941814 PMCID: PMC6164894 DOI: 10.3390/biomedicines6030074] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 12/29/2022] Open
Abstract
Fibrosis results from the excessive accumulation of extracellular matrix in chronically injured tissue. The fibrotic process is governed by crosstalk between many signaling pathways. The search for an effective treatment is further complicated by the fact that there is a degree of tissue-specificity in the pathways involved, although the process is not completely understood for all tissues. A plethora of drugs have shown promise in pre-clinical models, which is not always borne out translationally in clinical trial. With the recent approvals of two antisense oligonucleotides for the treatment of the genetic diseases Duchenne muscular dystrophy and spinal muscular atrophy, we explore here the potential of antisense oligonucleotides to knockdown the expression of pro-fibrotic proteins. We give an overview of the generalized fibrotic process, concentrating on key players and highlight where antisense oligonucleotides have been used effectively in cellular and animal models of different fibrotic conditions. Consideration is given to the advantages antisense oligonucleotides would have as an anti-fibrotic therapy alongside factors that would need to be addressed to improve efficacy. A prospective outlook for the development of antisense oligonucleotides to target fibrosis is outlined.
Collapse
Affiliation(s)
- James T March
- Centre for Gene and Cell Therapy, School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey TW20 0EX, UK.
| | - Golnoush Golshirazi
- Centre for Gene and Cell Therapy, School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey TW20 0EX, UK.
| | - Viktorija Cernisova
- Centre for Gene and Cell Therapy, School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey TW20 0EX, UK.
| | - Heidi Carr
- Centre for Gene and Cell Therapy, School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey TW20 0EX, UK.
| | - Yee Leong
- Centre for Gene and Cell Therapy, School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey TW20 0EX, UK.
| | - Ngoc Lu-Nguyen
- Centre for Gene and Cell Therapy, School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey TW20 0EX, UK.
| | - Linda J Popplewell
- Centre for Gene and Cell Therapy, School of Biological Sciences, Royal Holloway-University of London, Egham, Surrey TW20 0EX, UK.
| |
Collapse
|
22
|
Johnson RD, Camelliti P. Role of Non-Myocyte Gap Junctions and Connexin Hemichannels in Cardiovascular Health and Disease: Novel Therapeutic Targets? Int J Mol Sci 2018; 19:ijms19030866. [PMID: 29543751 PMCID: PMC5877727 DOI: 10.3390/ijms19030866] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 03/10/2018] [Accepted: 03/12/2018] [Indexed: 12/24/2022] Open
Abstract
The heart is a complex organ composed of multiple cell types, including cardiomyocytes and different non-myocyte populations, all working closely together to determine the hearts properties and maintain normal cardiac function. Connexins are abundantly expressed proteins that form plasma membrane hemichannels and gap junctions between cells. Gap junctions are intracellular channels that allow for communication between cells, and in the heart they play a crucial role in cardiac conduction by coupling adjacent cardiomyocytes. Connexins are expressed in both cardiomyocytes and non-myocytes, including cardiac fibroblasts, endothelial cells, and macrophages. Non-myocytes are the largest population of cells in the heart, and therefore it is important to consider what roles connexins, hemichannels, and gap junctions play in these cell types. The aim of this review is to provide insight into connexin-based signalling in non-myocytes during health and disease, and highlight how targeting these proteins could lead to the development of novel therapies. We conclude that connexins in non-myocytes contribute to arrhythmias and adverse ventricular remodelling following myocardial infarction, and are associated with the initiation and development of atherosclerosis. Therefore, therapeutic interventions targeting these connexins represent an exciting new research avenue with great potential.
Collapse
Affiliation(s)
- Robert D Johnson
- School of Biosciences and Medicine, University of Surrey, Guildford GU2 7XH, UK.
| | - Patrizia Camelliti
- School of Biosciences and Medicine, University of Surrey, Guildford GU2 7XH, UK.
| |
Collapse
|
23
|
Cho N, Razipour SE, McCain ML. Featured Article: TGF-β1 dominates extracellular matrix rigidity for inducing differentiation of human cardiac fibroblasts to myofibroblasts. Exp Biol Med (Maywood) 2018; 243:601-612. [PMID: 29504479 DOI: 10.1177/1535370218761628] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cardiac fibroblasts and their activated derivatives, myofibroblasts, play a critical role in wound healing after myocardial injury and often contribute to long-term pathological outcomes, such as excessive fibrosis. Thus, defining the microenvironmental factors that regulate the phenotype of cardiac fibroblasts and myofibroblasts could lead to new therapeutic strategies. Both chemical and biomechanical cues have previously been shown to induce myofibroblast differentiation in many organs and species. For example, transforming growth factor beta 1, a cytokine secreted by neutrophils, and rigid extracellular matrix environments have both been shown to promote differentiation. However, the relative contributions of transforming growth factor beta 1 and extracellular matrix rigidity, two hallmark cues in many pathological myocardial microenvironments, to the phenotype of human cardiac fibroblasts are unclear. We hypothesized that transforming growth factor beta 1 and rigid extracellular matrix environments would potentially have a synergistic effect on the differentiation of human cardiac fibroblasts to myofibroblasts. To test this, we seeded primary human adult cardiac fibroblasts onto coverslips coated with polydimethylsiloxane of various elastic moduli, introduced transforming growth factor beta 1, and longitudinally quantified cell phenotype by measuring expression of α-smooth muscle actin, the most robust indicator of myofibroblasts. Our data indicate that, although extracellular matrix rigidity influenced differentiation after one day of transforming growth factor beta 1 treatment, ultimately transforming growth factor beta 1 superseded extracellular matrix rigidity as the primary regulator of myofibroblast differentiation. We also measured expression of POSTN, FAP, and FSP1, proposed secondary indicators of fibroblast/myofibroblast phenotypes. Although these genes partially trended with α-smooth muscle actin expression, they were relatively inconsistent. Finally, we demonstrated that activated myofibroblasts incompletely revert to a fibroblast phenotype after they are re-plated onto new surfaces without transforming growth factor beta 1, suggesting differentiation is partially reversible. Our results provide new insights into how microenvironmental cues affect human cardiac fibroblast differentiation in the context of myocardial pathology, which is important for identifying effective therapeutic targets and dictating supporting cell phenotypes for engineered human cardiac disease models. Impact statement Heart disease is the leading cause of death worldwide. Many forms of heart disease are associated with fibrosis, which increases extracellular matrix (ECM) rigidity and compromises cardiac output. Fibrotic tissue is synthesized primarily by myofibroblasts differentiated from fibroblasts. Thus, defining the cues that regulate myofibroblast differentiation is important for understanding the mechanisms of fibrosis. However, previous studies have focused on non-human cardiac fibroblasts and have not tested combinations of chemical and mechanical cues. We tested the effects of TGF-β1, a cytokine secreted by immune cells after injury, and ECM rigidity on the differentiation of human cardiac fibroblasts to myofibroblasts. Our results indicate that differentiation is initially influenced by ECM rigidity, but is ultimately superseded by TGF-β1. This suggests that targeting TGF-β signaling pathways in cardiac fibroblasts may have therapeutic potential for attenuating fibrosis, even in rigid microenvironments. Additionally, our approach can be leveraged to engineer more precise multi-cellular human cardiac tissue models.
Collapse
Affiliation(s)
- Nathan Cho
- 1 Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Shadi E Razipour
- 1 Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Megan L McCain
- 1 Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.,2 Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
24
|
Coelho LL, Pereira IR, Pereira MCDS, Mesquita L, Lannes-Vieira J, Adesse D, Garzoni LR. Trypanosoma cruzi activates mouse cardiac fibroblasts in vitro leading to fibroblast-myofibroblast transition and increase in expression of extracellular matrix proteins. Parasit Vectors 2018; 11:72. [PMID: 29382361 PMCID: PMC5791182 DOI: 10.1186/s13071-018-2614-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 01/02/2018] [Indexed: 01/05/2023] Open
Abstract
Background Cardiac fibrosis is a consequence of chronic chagasic cardiomyopathy (CCC). In other cardiovascular diseases, the protagonist role of fibroblasts in cardiac fibrosis is well established. However, the role of cardiac fibroblasts (CFs) in fibrosis during the CCC is not clear. Here, our aim was to investigate the effect of Trypanosoma cruzi, the etiological agent of Chagas disease on CFs activation. Methods Cardiac fibroblasts were purified from primary cultures of mouse embryo cardiac cells. After two passages, cells were infected with T. cruzi (Y strain) and analyzed at different times for determination of infectivity, activation and production of extracellular matrix components (fibronectin, laminin and collagen IV) by immunofluorescence and western blot. Results At second passage, cultures were enriched in CFs (95% of fibroblasts and 5% of cardiomyocytes), as revealed by presence of alpha-smooth muscle actin (α-SMA) and discoidin domain receptor 2 (DDR2) and absence of sarcomeric tropomyosin (ST) protein expression. Trypanosoma cruzi infection induced fibroblast-myofibroblast transition, with increased expression of α-SMA after 6 and 24 h post-infection (hpi). Fibronectin was increased at 6, 24 and 48 hpi, laminin was increased at 6 and 24 hpi and collagen IV was increased at 6 hpi. Conclusions Our results showed that T. cruzi activates CFs, inducing activation and exacerbates ECM production. Furthermore, our data raise the possibility of the involvement of CFs in heart fibrosis during Chagas disease.
Collapse
Affiliation(s)
- Laura Lacerda Coelho
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Av. Brasil 4365, Pavilhão Cardoso Fontes, 2° andar, Rio de Janeiro RJ, 20045-900, Brazil
| | - Isabela Resende Pereira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Av. Brasil 4365, Pavilhão Cardoso Fontes, 2° andar, Rio de Janeiro RJ, 20045-900, Brazil
| | - Mirian Claudia de Souza Pereira
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Av. Brasil 4365, Pavilhão Carlos Chagas sala 308, Rio de Janeiro RJ, 20045-900, Brazil
| | - Liliane Mesquita
- Laboratório de Ultraestrutura Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Av. Brasil 4365, Pavilhão Carlos Chagas sala 308, Rio de Janeiro RJ, 20045-900, Brazil
| | - Joseli Lannes-Vieira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Av. Brasil 4365, Pavilhão Cardoso Fontes, 2° andar, Rio de Janeiro RJ, 20045-900, Brazil
| | - Daniel Adesse
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Av. Brasil 4365, Pavilhão Carlos Chagas, sala 307, Rio de Janeiro RJ, 20045-900, Brazil
| | - Luciana Ribeiro Garzoni
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Av. Brasil 4365, Pavilhão Cardoso Fontes, 2° andar, Rio de Janeiro RJ, 20045-900, Brazil.
| |
Collapse
|
25
|
Wu PC, Hsu WL, Chen CL, Lam CF, Huang YB, Huang CC, Lin MH, Lin MW. Morphine Induces Fibroblast Activation through Up-regulation of Connexin 43 Expression: Implication of Fibrosis in Wound Healing. Int J Med Sci 2018; 15:875-882. [PMID: 30008599 PMCID: PMC6036091 DOI: 10.7150/ijms.23074] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 02/02/2018] [Indexed: 12/15/2022] Open
Abstract
Morphine is the most effective drugs for attenuating various types of severe pain, but morphine abuse carries a high risk of systemic fibrosis. Our previous have indicated that systemic administration of morphine hinders angiogenesis and delays wound healing. Here we have explained the pathological mechanism underlying the effect of morphine on wound healing. To determine how morphine affects wound healing, we first created a wound in mice treated them with a combination of a low doses (5 mg/kg/day) and high doses (20 or 30 mg/kg/day) of morphine. An In vivo study revealed that high-dose morphine-induced abnormal myofibroblasts persist after the end of wound healing because of connexin 43 (Cx43) upregulation. High-dose morphine-induced Cx43 increased the expression levels of focal adhesion molecules, namely fibronectin and alpha-smooth muscle actin (α-SMA) through the activation of transforming growth factor (TGF)-β1 signaling. In addition, we found that Cx43 contributed to TGF-βRII/ Smad2/3 signaling for regulating the differentiation of fibroblasts into myofibroblasts during high-dose morphine exposure. In conclusion, the abnormal regulation of Cx43 by morphine may induce systemic fibrosis because of abnormal myofibroblast function.
Collapse
Affiliation(s)
- Ping-Ching Wu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan.,Institute of Oral Medicine and Department of Stomatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University Tainan, Taiwan.,Medical Device Innovation Center, Taiwan Innovation Center of Medical Devices and Technology, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Li Hsu
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chun-Lin Chen
- Department of Biological Science, National Sun Yat-sen University, Kaohsiung, Taiwan.,Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chen-Fuh Lam
- Department of Anesthesiology, E-Da Hospital/E-Da Cancer Hospital/I-Shou University, Kaohsiung, Taiwan
| | - Yaw-Bin Huang
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan.,School of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chien-Chi Huang
- Department of Anesthesiology, National Cheng Kung University College of Medicine and Hospital, Tainan, Taiwan
| | - Ming-Hong Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Ming-Wei Lin
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, E-Da Hospital/E-Da Cancer Hospital, Kaohsiung, Taiwan
| |
Collapse
|
26
|
Zhang WH, Li XL, Guo Y, Zhang Y. Proliferation and osteogenic activity of fibroblasts induced with fibronectin. ACTA ACUST UNITED AC 2017; 50:e6272. [PMID: 28832764 PMCID: PMC5561809 DOI: 10.1590/1414-431x20176272] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/22/2017] [Indexed: 11/25/2022]
Abstract
The aim of this study was to determine the proliferation and osteogenic activity of fibroblasts induced with fibronectin and their possible dose-dependent relationship. The fibroblasts obtained by tissue explants adherent method were induced with fibronectin at different concentrations of 0, 10, 20, 40, 60, and 80 μg/mL for 14 days. The 3H-thymidine and 3H-proline incorporation test was used to evaluate the synthesis of DNA and collagen by fibroblasts, respectively. The mineralized nodules and osteocalcin secretion, as vital osteogenic indicators, were detected with tetracycline labeling and 125I-labeled competitive immunoassay, respectively. Fibronectin significantly increased the synthesis of DNA and collagen by fibroblasts, especially at the concentration of 40 μg/mL (P<0.05). The increased secretion of osteocalcin in the supernatant was also statistically significant at the concentration of 40 μg/mL (P<0.05). The mineralized nodules with trabecula-like structure derived from induced fibroblasts were positive for tetracycline labeling. The granulation tissue-derived fibroblasts induced with fibronectin exhibited increased proliferative, functional and osteogenic potential. Fibroblasts are considered a possible in situ stem cell in tissue engineering.
Collapse
Affiliation(s)
| | - X-L Li
- Tianjin Hospital, Tianjin, China
| | - Y Guo
- Tianjin Hospital, Tianjin, China
| | - Y Zhang
- Tianjin Hospital, Tianjin, China
| |
Collapse
|
27
|
Paw M, Borek I, Wnuk D, Ryszawy D, Piwowarczyk K, Kmiotek K, Wójcik-Pszczoła KA, Pierzchalska M, Madeja Z, Sanak M, Błyszczuk P, Michalik M, Czyż J. Connexin43 Controls the Myofibroblastic Differentiation of Bronchial Fibroblasts from Patients with Asthma. Am J Respir Cell Mol Biol 2017; 57:100-110. [PMID: 28245135 DOI: 10.1165/rcmb.2015-0255oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pathologic accumulation of myofibroblasts in asthmatic bronchi is regulated by extrinsic stimuli and by the intrinsic susceptibility of bronchial fibroblasts to transforming growth factor-β (TGF-β). The specific function of gap junctions and connexins in this process has remained unknown. Here, we investigated the role of connexin43 (Cx43) in TGF-β-induced myofibroblastic differentiation of fibroblasts derived from bronchoscopic biopsy specimens of patients with asthma and donors without asthma. Asthmatic fibroblasts expressed considerably higher levels of Cx43 and were more susceptible to TGF-β1-induced myofibroblastic differentiation than were their nonasthmatic counterparts. TGF-β1 efficiently up-regulated Cx43 levels and activated the canonical Smad pathway in asthmatic cells. Ectopic Cx43 expression in nonasthmatic (Cx43low) fibroblasts increased their predilection to TGF-β1-induced Smad2 activation and fibroblast-myofibroblast transition. Transient Cx43 silencing in asthmatic (Cx43high) fibroblasts by Cx43 small interfering RNA attenuated the TGF-β1-triggered Smad2 activation and myofibroblast formation. Direct interactions of Smad2 and Cx43 with β-tubulin were demonstrated by co-immunoprecipitation assay, whereas the sensitivity of these interactions to TGF-β1 signaling was confirmed by Förster Resonance Energy Transfer analyses. Furthermore, inhibition of the TGF-β1/Smad pathway attenuated TGF-β1-triggered Cx43 up-regulation and myofibroblast differentiation of asthmatic fibroblasts. Chemical inhibition of gap junctional intercellular communication with 18 α-glycyrrhetinic acid did not affect the initiation of fibroblast-myofibroblast transition in asthmatic fibroblasts but interfered with the maintenance of their myofibroblastic phenotype. Collectively, our data identified Cx43 as a new player in the feedback mechanism regulating TGF-β1/Smad-dependent differentiation of bronchial fibroblasts. Thus, our observations point to Cx43 as a novel profibrotic factor in asthma progression.
Collapse
Affiliation(s)
- Milena Paw
- 1 Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Izabela Borek
- 1 Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Dawid Wnuk
- 1 Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Damian Ryszawy
- 1 Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Katarzyna Piwowarczyk
- 1 Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Katarzyna Kmiotek
- 1 Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Katarzyna A Wójcik-Pszczoła
- 1 Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.,2 Department of Medicine, Jagiellonian University Medical School, Kraków, Poland
| | | | - Zbigniew Madeja
- 1 Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Marek Sanak
- 2 Department of Medicine, Jagiellonian University Medical School, Kraków, Poland
| | - Przemysław Błyszczuk
- 3 Department of Food Biotechnology, Faculty of Food Technology, University of Agriculture, Kraków, Poland.,5 Department of Clinical Immunology, Institute of Pediatrics, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Marta Michalik
- 1 Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Jarosław Czyż
- 1 Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
28
|
Macfelda K, Kapeller B, Holly A, Podesser BK, Losert U, Brandes K, Goettel P, Mueller J. Bioelectrical signals improve cardiac function and modify gene expression of extracellular matrix components. ESC Heart Fail 2017; 4:291-300. [PMID: 28772035 PMCID: PMC5542736 DOI: 10.1002/ehf2.12169] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 04/14/2017] [Accepted: 05/30/2017] [Indexed: 11/10/2022] Open
Abstract
Aims Beyond the influence of stimulating devices on cardiac excitation, their use in treating patients with heart failure has positive effects on the myocardium at the molecular level. Electrical signals can induce a wide spectrum of effects in living tissue. Therefore, we sought to determine whether applying electrical microcurrent directly to failing hearts leads to functional improvement. Methods and results Sixteen male spontaneously hypertensive rats (SHRs) with heart failure underwent application of a patch electrode to the left ventricular epicardium and placement of a subcutaneous counter electrode. The electrode delivered a 0.35 μA microcurrent to nine of the SHRs for 45 ± 3 days; the other seven SHRs were used as controls. At baseline and before the SHRs were humanely put to death, we measured the left ventricular ejection fraction (LVEF) and the thickness of the LV posterior wall during systole and diastole (LVPWs/d). We used quantitative PCR to determine extracellular matrix parameters [collagen I–III, matrix metalloproteinase (MMP)‐2, MMP‐9, tissue inhibitor of metalloproteinases 3 (TIMP3), TIMP4, connexins (Cxs) 40/43/45, transforming growth factor (TGF)‐β, and interleukin (IL)‐6]. Among SHRs undergoing microcurrent application, LVEF normalized (mean decrease, 22.8%; P = 0.009), and LVPWs decreased (mean, 35.3%; P = 0.001). Compared with the control group, the SHRs receiving microcurrent exhibited a mean decrease in the gene expression of collagen I (10.6%, P = 0.003), TIMP3 (18.5%, P = 0.005), Cx43 (14.3%, P = 0.003), Cx45 (12.7%, P = 0.020), TGF‐β (13.0%, P = 0.005), and IL‐6 (53.7%, P = 0.000). Microcurrent application induced no changes in the expression of collagen III, MMP‐2, MMP‐9, TIMP4, or Cx40. Conclusions Applying microcurrent to the LV epicardium of SHRs leads to statistically significant functional improvement and alterations in the levels of inflammatory and extracellular matrix components.
Collapse
Affiliation(s)
- Karin Macfelda
- Department of Biomedical Research (Cell Biology), Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Barbara Kapeller
- Department of Biomedical Research (Cell Biology), Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Alexander Holly
- Department of Biomedical Research (Cell Biology), Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Bruno K Podesser
- Department of Biomedical Research (Cell Biology), Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Udo Losert
- Department of Biomedical Research (Cell Biology), Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Kersten Brandes
- Berlin Heals, Knesebeckstrasse 59-61, 10719, Berlin, Germany
| | - Peter Goettel
- Berlin Heals, Knesebeckstrasse 59-61, 10719, Berlin, Germany
| | | |
Collapse
|
29
|
Novel therapeutic strategies targeting fibroblasts and fibrosis in heart disease. Nat Rev Drug Discov 2016; 15:620-638. [PMID: 27339799 DOI: 10.1038/nrd.2016.89] [Citation(s) in RCA: 236] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Our understanding of the functions of cardiac fibroblasts has moved beyond their roles in heart structure and extracellular matrix generation and now includes their contributions to paracrine, mechanical and electrical signalling during ontogenesis and normal cardiac activity. Fibroblasts also have central roles in pathogenic remodelling during myocardial ischaemia, hypertension and heart failure. As key contributors to scar formation, they are crucial for tissue repair after interventions including surgery and ablation. Novel experimental approaches targeting cardiac fibroblasts are promising potential therapies for heart disease. Indeed, several existing drugs act, at least partially, through effects on cardiac connective tissue. This Review outlines the origins and roles of fibroblasts in cardiac development, homeostasis and disease; illustrates the involvement of fibroblasts in current and emerging clinical interventions; and identifies future targets for research and development.
Collapse
|
30
|
Ongstad EL, Gourdie RG. Can heart function lost to disease be regenerated by therapeutic targeting of cardiac scar tissue? Semin Cell Dev Biol 2016; 58:41-54. [PMID: 27234380 DOI: 10.1016/j.semcdb.2016.05.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/18/2016] [Accepted: 05/23/2016] [Indexed: 01/14/2023]
Abstract
Myocardial infarction results in scar tissue that cannot actively contribute to heart mechanical function and frequently causes lethal arrhythmias. The healing response after infarction involves inflammation, biochemical signaling, changes in cellular phenotype, activity, and organization, and alterations in electrical conduction due to variations in cell and tissue geometry and alterations in protein expression, organization, and function - particularly in membrane channels. The intensive research focus on regeneration of myocardial tissues has, as of yet, only met with modest success, with no near-term prospect of improving standard-of-care for patients with heart disease. An alternative concept for novel therapeutic approach is the rejuvenation of cardiac electrical and mechanical properties through the modification of scar tissue. Several peptide therapeutics, locally applied genetic therapies, or delivery of genetically modified cells have shown promise in improving the characteristics of the fibrous scar and post-myocardial infarction prognosis in experimental models. This review highlights several factors that contribute to arrhythmogenesis in scar formation and how these might be targeted to regenerate some of the electrical and mechanical function of the post-MI scar.
Collapse
Affiliation(s)
- Emily L Ongstad
- Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion Research Institute, 2 Riverside Circle, Roanoke, VA 24016, USA.
| | - Robert G Gourdie
- Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion Research Institute, 2 Riverside Circle, Roanoke, VA 24016, USA; Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, 317 Kelly Hall, Stanger Street, Blacksburg, VA 24061, USA; Department of Emergency Medicine, Carilion Clinic, 1906 Belleview Avenue, Roanoke VA 24014, USA.
| |
Collapse
|
31
|
Essa AAM, Yamazaki M, Maruyama S, Abé T, Babkair H, Raghib AM, Megahed EMED, Cheng J, Saku T. Tumour-associated macrophages are recruited and differentiated in the neoplastic stroma of oral squamous cell carcinoma. Pathology 2016; 48:219-27. [PMID: 27020496 DOI: 10.1016/j.pathol.2016.02.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 11/25/2015] [Accepted: 12/02/2015] [Indexed: 12/22/2022]
Abstract
To confirm our hypothesis that macrophages recruited to fight against oral squamous cell carcinoma (SCC) invasion are functionally differentiated within neoplastic stromata, we analysed arrangements of macrophage subtypes and cancer-associated fibroblasts (CAFs) in their association with blood vasculatures in the neoplastic stroma. Surgical specimens of oral SCC were immunohistochemically examined for macrophage phenotypes (CD68, CD163, and CD204) and stromal environments (perlecan, connexin 43, and CD31). Human monocytes were co-cultured with ZK-1 cells of oral SCC origin in different culture conditions. SCC stromata were divided into two types: fascicular (fibroblast-rich) and reticular (perlecan-rich). Regardless of stromal types, CD68 positive (+)/CD163+/CD204+ macrophages were recruited when blood vessels were abundant. Connexin 43+ fibroblasts were enriched in the fascicular stroma, where blood vessels were depleted. In co-culture experiments, monocytes, in the presence of ZK-1 cells, showed TNF-α(low)/IL-12(low) and IL-10(high)/VEGF(high)/MMP-9(high) with increased expression levels for fibronectin and perlecan. With direct contact with monocytes, SCC cells also expressed CD68 and CD163. SCC stromata were characterised by CD163+/CD204+ tumour-associated macrophages (TAMs) and connexin 43+ CAFs. TAMs are differentiated from monocytes by the physical contact with oral SCC cells in the perlecan-rich neoplastic stroma, which is also induced by the cross-talk between SCC cells and stromal cells including TAMs.
Collapse
Affiliation(s)
- Ahmed Abdelaziz Mohamed Essa
- Division of Oral Pathology, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Department of Oral Pathology, Faculty of Dentistry, Tanta University, Tanta, Egypt
| | - Manabu Yamazaki
- Division of Oral Pathology, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Satoshi Maruyama
- Oral Pathology Section, Department of Surgical Pathology, Niigata University Hospital, Niigata, Japan
| | - Tatsuya Abé
- Division of Oral Pathology, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Oral Pathology Section, Department of Surgical Pathology, Niigata University Hospital, Niigata, Japan
| | - Hamzah Babkair
- Division of Oral Pathology, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Adel Mohamed Raghib
- Department of Oral Pathology, Faculty of Dentistry, Tanta University, Tanta, Egypt
| | | | - Jun Cheng
- Division of Oral Pathology, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takashi Saku
- Division of Oral Pathology, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Oral Pathology Section, Department of Surgical Pathology, Niigata University Hospital, Niigata, Japan.
| |
Collapse
|
32
|
Cogliati B, Mennecier G, Willebrords J, Da Silva TC, Maes M, Pereira IVA, Crespo-Yanguas S, Hernandez-Blazquez FJ, Dagli MLZ, Vinken M. Connexins, Pannexins, and Their Channels in Fibroproliferative Diseases. J Membr Biol 2016; 249:199-213. [PMID: 26914707 DOI: 10.1007/s00232-016-9881-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/16/2016] [Indexed: 12/13/2022]
Abstract
Cellular and molecular mechanisms of wound healing, tissue repair, and fibrogenesis are established in different organs and are essential for the maintenance of function and tissue integrity after cell injury. These mechanisms are also involved in a plethora of fibroproliferative diseases or organ-specific fibrotic disorders, all of which are associated with the excessive deposition of extracellular matrix components. Fibroblasts, which are key cells in tissue repair and fibrogenesis, rely on communicative cellular networks to ensure efficient control of these processes and to prevent abnormal accumulation of extracellular matrix into the tissue. Despite the significant impact on human health, and thus the epidemiologic relevance, there is still no effective treatment for most fibrosis-related diseases. This paper provides an overview of current concepts and mechanisms involved in the participation of cellular communication via connexin-based pores as well as pannexin-based channels in the processes of tissue repair and fibrogenesis in chronic diseases. Understanding these mechanisms may contribute to the development of new therapeutic strategies to clinically manage fibroproliferative diseases and organ-specific fibrotic disorders.
Collapse
Affiliation(s)
- Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Brazil
| | - Gregory Mennecier
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Brazil
| | - Joost Willebrords
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Tereza Cristina Da Silva
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Brazil
| | - Michaël Maes
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Sara Crespo-Yanguas
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Maria Lúcia Zaidan Dagli
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Brazil
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
33
|
Ongstad E, Kohl P. Fibroblast-myocyte coupling in the heart: Potential relevance for therapeutic interventions. J Mol Cell Cardiol 2016; 91:238-46. [PMID: 26774702 DOI: 10.1016/j.yjmcc.2016.01.010] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 01/09/2016] [Accepted: 01/11/2016] [Indexed: 01/03/2023]
Abstract
Cardiac myocyte-fibroblast electrotonic coupling is a well-established fact in vitro. Indirect evidence of its presence in vivo exists, but few functional studies have been published. This review describes the current knowledge of fibroblast-myocyte electrical signaling in the heart. Further research is needed to understand the frequency and extent of heterocellular interactions in vivo in order to gain a better understanding of their relevance in healthy and diseased myocardium. It is hoped that associated insight into myocyte-fibroblast coupling in the heart may lead to the discovery of novel therapeutic targets and the development of agents for improving outcomes of myocardial scarring and fibrosis.
Collapse
Affiliation(s)
- Emily Ongstad
- Clemson University, Department of Bioengineering, Clemson, SC, USA; Virginia Tech Carilion Research Institute, Roanoke, VA, USA.
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg - Bad Krozingen, Faculty of Medicine, University Freiburg, Germany; Cardiac Biophysics and Systems Biology, National Heart and Lung Institute, Imperial College London, UK
| |
Collapse
|
34
|
Calcium signaling and the novel anti-proliferative effect of the UTP-sensitive P2Y11 receptor in rat cardiac myofibroblasts. Cell Calcium 2015; 58:518-33. [PMID: 26324417 DOI: 10.1016/j.ceca.2015.08.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 08/06/2015] [Accepted: 08/14/2015] [Indexed: 12/16/2022]
Abstract
During myocardial ischemia and reperfusion both purines and pyrimidines are released into the extracellular milieu, thus creating a signaling wave that propagates to neighboring cells via membrane-bound P2 purinoceptors activation. Cardiac fibroblasts (CF) are important players in heart remodeling, electrophysiological changes and hemodynamic alterations following myocardial infarction. Here, we investigated the role UTP on calcium signaling and proliferation of CF cultured from ventricles of adult rats. Co-expression of discoidin domain receptor 2 and α-smooth muscle actin indicate that cultured CF are activated myofibroblasts. Intracellular calcium ([Ca(2+)]i) signals were monitored in cells loaded with Fluo-4 NW. CF proliferation was evaluated by the MTT assay. UTP and the selective P2Y4 agonist, MRS4062, caused a fast desensitizing [Ca(2+)]i rise originated from thapsigargin-sensitive internal stores, which partially declined to a plateau providing the existence of Ca(2+) in the extracellular fluid. The biphasic [Ca(2+)]i response to UTP was attenuated respectively by P2Y4 blockers, like reactive blue-2 and suramin, and by the P2Y11 antagonist, NF340. UTP and the P2Y2 receptor agonist MRS2768 increased, whereas the selective P2Y11 agonist NF546 decreased, CF growth; MRS4062 was ineffective. Blockage of the P2Y11 receptor or its coupling to adenylate cyclase boosted UTP-induced CF proliferation. Confocal microscopy and Western blot analysis confirmed the presence of P2Y2, P2Y4 and P2Y11 receptors. Data indicate that besides P2Y4 and P2Y2 receptors which are responsible for UTP-induced [Ca(2+)]i transients and growth of CF, respectively, synchronous activation of the previously unrecognized P2Y11 receptor may represent an important target for anti-fibrotic intervention in cardiac remodeling.
Collapse
|
35
|
Atrial Fibrillation and Fibrosis: Beyond the Cardiomyocyte Centric View. BIOMED RESEARCH INTERNATIONAL 2015; 2015:798768. [PMID: 26229964 PMCID: PMC4502285 DOI: 10.1155/2015/798768] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 03/27/2015] [Accepted: 03/30/2015] [Indexed: 12/31/2022]
Abstract
Atrial fibrillation (AF) associated with fibrosis is characterized by the appearance of interstitial myofibroblasts. These cells are responsible for the uncontrolled deposition of the extracellular matrix, which pathologically separate cardiomyocyte bundles. The enhanced fibrosis is thought to contribute to arrhythmias “indirectly” because a collagenous septum is a passive substrate for propagation, resulting in impulse conduction block and/or zigzag conduction. However, the emerging results demonstrate that myofibroblasts in vitro also promote arrhythmogenesis due to direct implications upon cardiomyocyte electrophysiology. This electrical interference may be considered beneficial as it resolves any conduction blocks; however, the passive properties of myofibroblasts might cause a delay in impulse propagation, thus promoting AF due to discontinuous slow conduction. Moreover, low-polarized myofibroblasts reduce, via cell-density dependence, the fast driving inward current for cardiac impulse conduction, therefore resulting in arrhythmogenic uniformly slow propagation. Critically, the subsequent reduction in cardiomyocytes resting membrane potential in vitro significantly increases the likelihood of ectopic activity. Myofibroblast densities and the degree of coupling at cellular border zones also impact upon this likelihood. By considering future in vivo studies, which identify myofibroblasts “per se” as a novel targets for cardiac arrhythmias, this review aims to describe the implications of noncardiomyocyte view in the context of AF.
Collapse
|
36
|
Balla P, Maros ME, Barna G, Antal I, Papp G, Sapi Z, Athanasou NA, Benassi MS, Picci P, Krenacs T. Prognostic impact of reduced connexin43 expression and gap junction coupling of neoplastic stromal cells in giant cell tumor of bone. PLoS One 2015; 10:e0125316. [PMID: 25933380 PMCID: PMC4416750 DOI: 10.1371/journal.pone.0125316] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 03/12/2015] [Indexed: 12/05/2022] Open
Abstract
Missense mutations of the GJA1 gene encoding the gap junction channel protein connexin43 (Cx43) cause bone malformations resulting in oculodentodigital dysplasia (ODDD), while GJA1 null and ODDD mutant mice develop osteopenia. In this study we investigated Cx43 expression and channel functions in giant cell tumor of bone (GCTB), a locally aggressive osteolytic lesion with uncertain progression. Cx43 protein levels assessed by immunohistochemistry were correlated with GCTB cell types, clinico-radiological stages and progression free survival in tissue microarrays of 89 primary and 34 recurrent GCTB cases. Cx43 expression, phosphorylation, subcellular distribution and gap junction coupling was also investigated and compared between cultured neoplastic GCTB stromal cells and bone marow stromal cells or HDFa fibroblasts as a control. In GCTB tissues, most Cx43 was produced by CD163 negative neoplastic stromal cells and less by CD163 positive reactive monocytes/macrophages or by giant cells. Significantly less Cx43 was detected in α-smooth muscle actin positive than α-smooth muscle actin negative stromal cells and in osteoclast-rich tumor nests than in the adjacent reactive stroma. Progressively reduced Cx43 production in GCTB was significantly linked to advanced clinico-radiological stages and worse progression free survival. In neoplastic GCTB stromal cell cultures most Cx43 protein was localized in the paranuclear-Golgi region, while it was concentrated in the cell membranes both in bone marrow stromal cells and HDFa fibroblasts. In Western blots, alkaline phosphatase sensitive bands, linked to serine residues (Ser369, Ser372 or Ser373) detected in control cells, were missing in GCTB stromal cells. Defective cell membrane localization of Cx43 channels was in line with the significantly reduced transfer of the 622 Da fluorescing calcein dye between GCTB stromal cells. Our results show that significant downregulation of Cx43 expression and gap junction coupling in neoplastic stromal cells are associated with the clinical progression and worse prognosis in GCTB.
Collapse
MESH Headings
- Actins/genetics
- Actins/metabolism
- Adolescent
- Adult
- Aged
- Alkaline Phosphatase/deficiency
- Alkaline Phosphatase/genetics
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Bone Marrow Cells/metabolism
- Bone Marrow Cells/pathology
- Bone Neoplasms/diagnosis
- Bone Neoplasms/genetics
- Bone Neoplasms/metabolism
- Bone Neoplasms/pathology
- Bone and Bones/metabolism
- Bone and Bones/pathology
- Child
- Child, Preschool
- Connexin 43/genetics
- Connexin 43/metabolism
- Gap Junctions/metabolism
- Gap Junctions/pathology
- Gene Expression Regulation, Neoplastic
- Giant Cell Tumor of Bone/diagnosis
- Giant Cell Tumor of Bone/genetics
- Giant Cell Tumor of Bone/metabolism
- Giant Cell Tumor of Bone/pathology
- Giant Cells/metabolism
- Giant Cells/pathology
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Humans
- Macrophages/metabolism
- Macrophages/pathology
- Middle Aged
- Monocytes/metabolism
- Monocytes/pathology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Primary Cell Culture
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Signal Transduction
- Survival Analysis
Collapse
Affiliation(s)
- Peter Balla
- 1st Department of Pathology & Experimental Cancer Research, Semmelweis University Budapest, Hungary
| | - Mate Elod Maros
- 1st Department of Pathology & Experimental Cancer Research, Semmelweis University Budapest, Hungary
- Department of Neuroradiology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Gabor Barna
- 1st Department of Pathology & Experimental Cancer Research, Semmelweis University Budapest, Hungary
| | - Imre Antal
- Department of Orthopaedics, Semmelweis University, Budapest, Hungary
| | - Gergo Papp
- 1st Department of Pathology & Experimental Cancer Research, Semmelweis University Budapest, Hungary
| | - Zoltan Sapi
- 1st Department of Pathology & Experimental Cancer Research, Semmelweis University Budapest, Hungary
| | | | - Maria Serena Benassi
- Laboratory of Experimental Oncology, Institute of Orthopaedics Rizzoli, Bologna, Italy
| | - Pierro Picci
- Laboratory of Experimental Oncology, Institute of Orthopaedics Rizzoli, Bologna, Italy
| | - Tibor Krenacs
- 1st Department of Pathology & Experimental Cancer Research, Semmelweis University Budapest, Hungary
- Hunragian Academy of Sciences-Semmelweis University (MTA-SE) Tumor Progression Research Group, Budapest, Hungary
- * E-mail:
| |
Collapse
|
37
|
Tarzemany R, Jiang G, Larjava H, Häkkinen L. Expression and function of connexin 43 in human gingival wound healing and fibroblasts. PLoS One 2015; 10:e0115524. [PMID: 25584940 PMCID: PMC4293150 DOI: 10.1371/journal.pone.0115524] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 11/25/2014] [Indexed: 11/18/2022] Open
Abstract
Connexins (C×s) are a family of transmembrane proteins that form hemichannels and gap junctions (GJs) on the cell membranes, and transfer small signaling molecules between the cytoplasm and extracellular space and between connecting cells, respectively. Among C×s, suppressing C×43 expression or function promotes skin wound closure and granulation tissue formation, and may alleviate scarring, but the mechanisms are not well understood. Oral mucosal gingiva is characterized by faster wound closure and scarless wound healing outcome as compared to skin wounds. Therefore, we hypothesized that C×43 function is down regulated during human gingival wound healing, which in fibroblasts promotes expression of genes conducive for fast and scarless wound healing. Cultured gingival fibroblasts expressed C×43 as their major connexin. Immunostaining of unwounded human gingiva showed that C×43 was abundantly present in the epithelium, and in connective tissue formed large C×43 plaques in fibroblasts. At the early stages of wound healing, C×43 was strongly down regulated in wound epithelial cells and fibroblasts, returning to the level of normal tissue by day 60 post-wounding. Blocking of C×43 function by C×43 mimetic peptide Gap27 suppressed GJ-mediated dye transfer, promoted migration, and caused significant changes in the expression of wound healing-associated genes in gingival fibroblasts. In particular, out of 54 genes analyzed, several MMPs and TGF-β1, involved in regulation of inflammation and extracellular matrix (ECM) turnover, and VEGF-A, involved in angiogenesis, were significantly upregulated while pro-fibrotic ECM molecules, including Collagen type I, and cell contractility-related molecules were significantly down regulated. These responses involved MAPK, GSK3α/β and TGF-β signaling pathways, and AP1 and SP1 transcription factors. Thus, suppressed function of C×43 in fibroblasts promotes their migration, and regulates expression of wound healing-associated genes via AP1, SP1, MAPK, GSK3α/β and TGF-β signaling pathways, and may promote fast and scarless wound healing in human gingiva.
Collapse
Affiliation(s)
- Rana Tarzemany
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Guoqiao Jiang
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Hannu Larjava
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Lari Häkkinen
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
- * E-mail:
| |
Collapse
|
38
|
Combining TGF-β signal inhibition and connexin43 silencing for iPSC induction from mouse cardiomyocytes. Sci Rep 2014; 4:7323. [PMID: 25471520 PMCID: PMC4255192 DOI: 10.1038/srep07323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 11/18/2014] [Indexed: 01/17/2023] Open
Abstract
The reprogramming of differentiated cells into induced pluripotent stem cells (iPSCs) can be achieved by ectopic expression of defined transcription factors (Oct3/4, Sox2, Klf4 and c-Myc). However, to date, some iPSCs have been generated using viral vectors; thus, unexpected insertional mutagenesis in the target cells would be a potential risk. Here we report reprogramming of siPSCs (gene silencing-induced pluripotent stem cells) from mouse neonatal cardiomyocytes (CMs) by combining TGF-β signal inhibition and connexin43 (Cx43) silencing, and show that siPSCs show pluripotency in vitro and in vivo. Our novel non-insertional mutagenesis technique may provide a means for iPSC generation.
Collapse
|
39
|
Pinney JR, Du KT, Ayala P, Fang Q, Sievers RE, Chew P, Delrosario L, Lee RJ, Desai TA. Discrete microstructural cues for the attenuation of fibrosis following myocardial infarction. Biomaterials 2014; 35:8820-8828. [PMID: 25047625 DOI: 10.1016/j.biomaterials.2014.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 07/02/2014] [Indexed: 01/14/2023]
Abstract
Chronic fibrosis caused by acute myocardial infarction (MI) leads to increased morbidity and mortality due to cardiac dysfunction. We have developed a therapeutic materials strategy that aims to mitigate myocardial fibrosis by utilizing injectable polymeric microstructures to mechanically alter the microenvironment. Polymeric microstructures were fabricated using photolithographic techniques and studied in a three-dimensional culture model of the fibrotic environment and by direct injection into the infarct zone of adult rats. Here, we show dose-dependent down-regulation of expression of genes associated with the mechanical fibrotic response in the presence of microstructures. Injection of this microstructured material into the infarct zone decreased levels of collagen and TGF-β, increased elastin deposition and vascularization in the infarcted region, and improved functional outcomes after six weeks. Our results demonstrate the efficacy of these discrete anti-fibrotic microstructures and suggest a potential therapeutic materials approach for combatting pathologic fibrosis.
Collapse
Affiliation(s)
- James R Pinney
- UC Berkeley - UCSF Graduate Group in Bioengineering, 1700 4th Street, QB3 Byers Hall, Room 203, San Francisco, CA 94158, USA; UCSF Medical Scientist Training Program, 1700 4th Street, QB3 Byers Hall, Room 203, San Francisco, CA 94158, USA
| | - Kim T Du
- UCSF Department of Medicine, Cardiovascular Research Institute and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Box 1354, 513 Parnassus Ave, MS Room 1136, San Francisco, CA 94143, USA
| | - Perla Ayala
- UC Berkeley - UCSF Graduate Group in Bioengineering, 1700 4th Street, QB3 Byers Hall, Room 203, San Francisco, CA 94158, USA; Beth Israel Deaconess Medical Center, Department of Surgery, Center for Life Science Surgery/BIDMC, 11th Floor, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Qizhi Fang
- UCSF Department of Medicine, Cardiovascular Research Institute and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Box 1354, 513 Parnassus Ave, MS Room 1136, San Francisco, CA 94143, USA
| | - Richard E Sievers
- UCSF Department of Medicine, Cardiovascular Research Institute and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Box 1354, 513 Parnassus Ave, MS Room 1136, San Francisco, CA 94143, USA
| | - Patrick Chew
- UCSF Bioengineering and Therapeutic Sciences, 1700 4th Street, Byers Hall Room 203, San Francisco, CA 94158, USA
| | - Lawrence Delrosario
- UCSF School of Medicine, 513 Parnassus Ave, MS Room 1136, San Francisco, CA 94143, USA
| | - Randall J Lee
- UC Berkeley - UCSF Graduate Group in Bioengineering, 1700 4th Street, QB3 Byers Hall, Room 203, San Francisco, CA 94158, USA; UCSF Department of Medicine, Cardiovascular Research Institute and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Box 1354, 513 Parnassus Ave, MS Room 1136, San Francisco, CA 94143, USA
| | - Tejal A Desai
- UC Berkeley - UCSF Graduate Group in Bioengineering, 1700 4th Street, QB3 Byers Hall, Room 203, San Francisco, CA 94158, USA; UCSF Bioengineering and Therapeutic Sciences, 1700 4th Street, Byers Hall Room 203, San Francisco, CA 94158, USA.
| |
Collapse
|
40
|
Abed A, Toubas J, Kavvadas P, Authier F, Cathelin D, Alfieri C, Boffa JJ, Dussaule JC, Chatziantoniou C, Chadjichristos CE. Targeting connexin 43 protects against the progression of experimental chronic kidney disease in mice. Kidney Int 2014; 86:768-79. [PMID: 24850151 DOI: 10.1038/ki.2014.108] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 02/17/2014] [Accepted: 02/20/2014] [Indexed: 11/09/2022]
Abstract
Excessive recruitment of monocytes and progression of fibrosis are hallmarks of chronic kidney disease (CKD). Recently we reported that the expression of connexin 43 (Cx43) was upregulated in the kidney during experimental nephropathy. To investigate the role of Cx43 in the progression of CKD, we interbred RenTg mice, a genetic model of hypertension-induced CKD, with Cx43+/- mice. The renal cortex of 5-month-old RenTgCx43+/- mice showed a marked decrease of cell adhesion markers leading to reduced monocyte infiltration and interstitial renal fibrosis compared with their littermates. In addition, functional and histological parameters such as albuminuria and glomerulosclerosis were ameliorated in RenTgCx43+/- mice. Interestingly, treatment with Cx43 antisense produced remarkable improvement of renal function and structure in 1-year-old RenTg mice. Similar results were found in Cx43+/- or wild-type mice treated with Cx43 antisense after obstructive nephropathy. Furthermore, in these mice, Cx43 antisense attenuated E-cadherin downregulation and phosphorylation of the transcription factor Sp1 by the ERK pathway resulting in decreased transcription of type I collagen gene. Interestingly, Cx43-specific blocking peptide inhibited monocyte adhesion in activated endothelium and profibrotic pathways in tubular cells. Cx43 was highly increased in biopsies of patients with CKD. Thus, Cx43 may represent a new therapeutic target against the progression of CKD.
Collapse
Affiliation(s)
- Ahmed Abed
- 1] INSERM UMR-S1155, Tenon Hospital, Paris, France [2] Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| | - Julie Toubas
- 1] INSERM UMR-S1155, Tenon Hospital, Paris, France [2] Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| | | | | | | | | | - Jean-Jacques Boffa
- 1] INSERM UMR-S1155, Tenon Hospital, Paris, France [2] Sorbonne Universités, UPMC Univ Paris 6, Paris, France [3] Department of Nephrology, Tenon Hospital, Paris, France
| | - Jean-Claude Dussaule
- 1] INSERM UMR-S1155, Tenon Hospital, Paris, France [2] Sorbonne Universités, UPMC Univ Paris 6, Paris, France [3] Department of Physiology, Saint-Antoine Hospital, Paris, France
| | - Christos Chatziantoniou
- 1] INSERM UMR-S1155, Tenon Hospital, Paris, France [2] Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| | - Christos E Chadjichristos
- 1] INSERM UMR-S1155, Tenon Hospital, Paris, France [2] Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| |
Collapse
|
41
|
Grek CL, Rhett JM, Ghatnekar GS. Cardiac to cancer: connecting connexins to clinical opportunity. FEBS Lett 2014; 588:1349-64. [PMID: 24607540 DOI: 10.1016/j.febslet.2014.02.047] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 02/25/2014] [Accepted: 02/26/2014] [Indexed: 12/26/2022]
Abstract
Gap junctions and their connexin components are indispensable in mediating the cellular coordination required for tissue and organ homeostasis. The critical nature of their existence mandates a connection to disease while at the same time offering therapeutic potential. Therapeutic intervention may be offered through the pharmacological and molecular disruption of the pathways involved in connexin biosynthesis, gap junction assembly, stabilization, or degradation. Chemical inhibitors aimed at closing connexin channels, peptide mimetics corresponding to short connexin sequences, and gene therapy approaches have been incredibly useful molecular tools in deciphering the complexities associated with connexin biology. Recently, therapeutic potential in targeting connexins has evolved from basic research in cell-based models to clinical opportunity in the form of human trials. Clinical promise is particularly evident with regards to targeting connexin43 in the context of wound healing. The following review is aimed at highlighting novel advances where the pharmacological manipulation of connexin biology has proven beneficial in animals or humans.
Collapse
Affiliation(s)
- Christina L Grek
- FirstString Research, Inc., 300 W. Coleman Blvd., Suite 203, Mount Pleasant, SC, United States
| | - J Matthew Rhett
- Department of Surgery, Division of General Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Gautam S Ghatnekar
- FirstString Research, Inc., 300 W. Coleman Blvd., Suite 203, Mount Pleasant, SC, United States.
| |
Collapse
|
42
|
Isometric Contraction of Dupuytren’s Myofibroblasts Is Inhibited by Blocking Intercellular Junctions. J Invest Dermatol 2013; 133:2664-2671. [DOI: 10.1038/jid.2013.219] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 04/09/2013] [Accepted: 04/09/2013] [Indexed: 01/22/2023]
|
43
|
Pinheiro AR, Paramos-de-Carvalho D, Certal M, Costa C, Magalhães-Cardoso MT, Ferreirinha F, Costa MA, Correia-de-Sá P. Bradykinin-induced Ca2+ signaling in human subcutaneous fibroblasts involves ATP release via hemichannels leading to P2Y12 receptors activation. Cell Commun Signal 2013; 11:70. [PMID: 24047499 PMCID: PMC3848849 DOI: 10.1186/1478-811x-11-70] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 09/12/2013] [Indexed: 12/31/2022] Open
Abstract
Background Chronic musculoskeletal pain involves connective tissue remodeling triggered by inflammatory mediators, such as bradykinin. Fibroblast cells signaling involve changes in intracellular Ca2+ ([Ca2+]i). ATP has been related to connective tissue mechanotransduction, remodeling and chronic inflammatory pain, via P2 purinoceptors activation. Here, we investigated the involvement of ATP in bradykinin-induced Ca2+ signals in human subcutaneous fibroblasts. Results Bradykinin, via B2 receptors, caused an abrupt rise in [Ca2+]i to a peak that declined to a plateau, which concentration remained constant until washout. The plateau phase was absent in Ca2+-free medium; [Ca2+]i signal was substantially reduced after depleting intracellular Ca2+ stores with thapsigargin. Extracellular ATP inactivation with apyrase decreased the [Ca2+]i plateau. Human subcutaneous fibroblasts respond to bradykinin by releasing ATP via connexin and pannexin hemichannels, since blockade of connexins, with 2-octanol or carbenoxolone, and pannexin-1, with 10Panx, attenuated bradykinin-induced [Ca2+]i plateau, whereas inhibitors of vesicular exocytosis, such as brefeldin A and bafilomycin A1, were inactive. The kinetics of extracellular ATP catabolism favors ADP accumulation in human fibroblast cultures. Inhibition of ectonucleotidase activity and, thus, ADP formation from released ATP with POM-1 or by Mg2+ removal from media reduced bradykinin-induced [Ca2+]i plateau. Selective blockade of the ADP-sensitive P2Y12 receptor with AR-C66096 attenuated bradykinin [Ca2+]i plateau, whereas the P2Y1 and P2Y13 receptor antagonists, respectively MRS 2179 and MRS 2211, were inactive. Human fibroblasts exhibited immunoreactivity against connexin-43, pannexin-1 and P2Y12 receptor. Conclusions Bradykinin induces ATP release from human subcutaneous fibroblasts via connexin and pannexin-1-containing hemichannels leading to [Ca2+]i mobilization through the cooperation of B2 and P2Y12 receptors.
Collapse
Affiliation(s)
- Ana Rita Pinheiro
- Laboratório de Farmacologia e Neurobiologia, Unidade Multidisciplinar de Investigação Biomédica (UMIB), Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, Edif, 2 Piso 4, Porto 4050-313, Portugal.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Pinheiro AR, Paramos-de-Carvalho D, Certal M, Costa MA, Costa C, Magalhães-Cardoso MT, Ferreirinha F, Sévigny J, Correia-de-Sá P. Histamine induces ATP release from human subcutaneous fibroblasts, via pannexin-1 hemichannels, leading to Ca2+ mobilization and cell proliferation. J Biol Chem 2013; 288:27571-27583. [PMID: 23918924 DOI: 10.1074/jbc.m113.460865] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Changes in the regulation of connective tissue ATP-mediated mechano-transduction and remodeling may be an important link to the pathogenesis of chronic pain. It has been demonstrated that mast cell-derived histamine plays an important role in painful fibrotic diseases. Here we analyzed the involvement of ATP in the response of human subcutaneous fibroblasts to histamine. Acute histamine application caused a rise in intracellular Ca(2+) ([Ca(2+)]i) and ATP release from human subcutaneous fibroblasts via H1 receptor activation. Histamine-induced [Ca(2+)]i rise was partially attenuated by apyrase, an enzyme that inactivates extracellular ATP, and by blocking P2 purinoceptors with pyridoxal phosphate-6-azo(benzene-2,4-disulfonic acid) tetrasodium salt and reactive blue 2. [Ca(2+)]i accumulation caused by histamine was also reduced upon blocking pannexin-1 hemichannels with (10)Panx, probenecid, or carbenoxolone but not when connexin hemichannels were inhibited with mefloquine or 2-octanol. Brefeldin A, an inhibitor of vesicular exocytosis, also did not block histamine-induced [Ca(2+)]i mobilization. Prolonged exposure of human subcutaneous fibroblast cultures to histamine favored cell growth and type I collagen synthesis via the activation of H1 receptor. This effect was mimicked by ATP and its metabolite, ADP, whereas the selective P2Y1 receptor antagonist, MRS2179, partially attenuated histamine-induced cell growth and type I collagen production. Expression of pannexin-1 and ADP-sensitive P2Y1 receptor on human subcutaneous fibroblasts was confirmed by immunofluorescence confocal microscopy and Western blot analysis. In conclusion, histamine induces ATP release from human subcutaneous fibroblasts, via pannexin-1 hemichannels, leading to [Ca(2+)]i mobilization and cell growth through the cooperation of H1 and P2 (probably P2Y1) receptors.
Collapse
Affiliation(s)
- Ana Rita Pinheiro
- Laboratório de Farmacologia e Neurobiologia, Unidade Multidisciplinar de Investigação Biomédica (UMIB); Área Técnico-Científica de Fisioterapia, Escola Superior de Tecnologia da Saúde do Instituto Politécnico do Porto, 4400-330 Vila Nova de Gaia, Portugal
| | - Diogo Paramos-de-Carvalho
- Laboratório de Farmacologia e Neurobiologia, Unidade Multidisciplinar de Investigação Biomédica (UMIB)
| | - Mariana Certal
- Laboratório de Farmacologia e Neurobiologia, Unidade Multidisciplinar de Investigação Biomédica (UMIB)
| | - Maria Adelina Costa
- Laboratório de Farmacologia e Neurobiologia, Unidade Multidisciplinar de Investigação Biomédica (UMIB); Departamento de Química, Instituto de Ciências Biomédicas Abel Salazar-Universidade do Porto (ICBAS-UP), 4050-313 Porto, Portugal
| | - Cristina Costa
- Laboratório de Farmacologia e Neurobiologia, Unidade Multidisciplinar de Investigação Biomédica (UMIB)
| | | | - Fátima Ferreirinha
- Laboratório de Farmacologia e Neurobiologia, Unidade Multidisciplinar de Investigação Biomédica (UMIB)
| | - Jean Sévigny
- Centre de Recherche en Rhumatologie et Immunologie, Centre Hospitalier Universitaire de Québec, Québec, Québec G1V 4G2, Canada; Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine, Université Laval, Québec, Québec G1V 0A6, Canada
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, Unidade Multidisciplinar de Investigação Biomédica (UMIB).
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Interest in the myofibroblast as a key player in propagation of chronic progressive fibrosis continues to elicit many publications, with focus on its cellular origins and the mechanisms underpinning their differentiation and/or transition. The objective of the review is to highlight this recent progress. RECENT FINDINGS The epithelial origin of the myofibroblast in fibrosis has been challenged by recent studies, with the pericyte suggested as a possible precursor instead. Additional signaling pathways, including Notch, Wnt, and hedgehog, are implicated in myofibroblast differentiation. The importance of NADPH oxidase 4 was highlighted recently to suggest a potential link between cellular/oxidative stress and the genesis of the myofibroblast. Recent observations on the importance of lysophosphatidic acid in fibrosis suggest that this may be due, in part, to its ability to regulate myofibroblast differentiation. Finally, there is increasing evidence for the role of epigenetic mechanisms in regulating myofibroblast differentiation, including DNA methylation and miRNA regulation of gene expression. SUMMARY These recent discoveries open up a whole new array of potential targets for novel antifibrotic therapies. This is of special importance given the current bleak outlook for chronic progressive fibrotic diseases, such as scleroderma, due to lack of effective therapies.
Collapse
|
46
|
Abstract
OBJECTIVES Pancreatic stellate cells (PSCs) play a pivotal role in pancreatic fibrosis associated with chronic pancreatitis and pancreatic cancer. Connexins (Cxs) allow direct intercellular communications as components of gap junction but also play important roles in the regulation of cell proliferation, cell differentiation, and tissue development. We here examined the expression of Cxs and Cx-mediated regulation of cell functions in PSCs. METHODS Human PSCs were isolated from patients undergoing operation for chronic pancreatitis or pancreatic cancer. The expression of Cxs was examined by reverse transcription polymerase chain reaction, Western blotting, and immunofluorescent staining. The roles of Cxs in PSC functions were examined by using carbenoxolone, a broad-spectrum Cx inhibitor, and small interfering RNA for Cx43. RESULTS Human activated PSCs expressed a variety of Cxs including Cx43 both in vitro and in vivo. Carbenoxolone inhibited platelet-derived growth factor-BB-induced proliferation and migration, and type I collagen expression in PSCs. In addition, carbenoxolone inhibited the activation of quiescent PSCs to a myofibroblastlike phenotype. Decreased Cx43 expression by small interfering RNA resulted in decreased proliferation and type I collagen expression. CONCLUSIONS Pancreatic stellate cells expressed a variety of Cxs. Connexins, especially Cx43, might regulate the cell functions and activation of PSCs.
Collapse
|
47
|
Häkkinen L, Larjava H, Koivisto L. Granulation tissue formation and remodeling. ACTA ACUST UNITED AC 2012. [DOI: 10.1111/etp.12008] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
48
|
Zhang P, Su J, Mende U. Cross talk between cardiac myocytes and fibroblasts: from multiscale investigative approaches to mechanisms and functional consequences. Am J Physiol Heart Circ Physiol 2012; 303:H1385-96. [PMID: 23064834 DOI: 10.1152/ajpheart.01167.2011] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The heart is comprised of a syncytium of cardiac myocytes (CM) and surrounding nonmyocytes, the majority of which are cardiac fibroblasts (CF). CM and CF are highly interspersed in the myocardium with one CM being surrounded by one or more CF. Bidirectional cross talk between CM and CF plays important roles in determining cardiac mechanical and electrical function in both normal and diseased hearts. Genetically engineered animal models and in vitro studies have provided evidence that CM and CF can regulate each other's function. Their cross talk contributes to structural and electrical remodeling in both atria and ventricles and appears to be involved in the pathogenesis of various heart diseases that lead to heart failure and arrhythmia disorders. Mechanisms of CM-CF cross talk, which are not yet fully understood, include release of paracrine factors, direct cell-cell interactions via gap junctions and potentially adherens junctions and nanotubes, and cell interactions with the extracellular matrix. In this article, we provide an overview of the existing multiscale experimental and computational approaches for the investigation of cross talk between CM and CF and review recent progress in our understanding of the functional consequences and underlying mechanisms. Targeting cross talk between CM and CF could potentially be used therapeutically for the modulation of the cardiac remodeling response in the diseased heart and may lead to new strategies for the treatment of heart failure or rhythm disturbances.
Collapse
Affiliation(s)
- P Zhang
- Cardiovascular Research Center, Cardiology Division, Rhode Island Hospital, Providence, USA
| | | | | |
Collapse
|
49
|
Forte E, Miraldi F, Chimenti I, Angelini F, Zeuner A, Giacomello A, Mercola M, Messina E. TGFβ-dependent epithelial-to-mesenchymal transition is required to generate cardiospheres from human adult heart biopsies. Stem Cells Dev 2012; 21:3081-90. [PMID: 22765842 DOI: 10.1089/scd.2012.0277] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Autologous cardiac progenitor cells (CPCs) isolated as cardiospheres (CSps) represent a promising candidate for cardiac regenerative therapy. A better understanding of the origin and mechanisms underlying human CSps formation and maturation is undoubtedly required to enhance their cardiomyogenic potential. Epithelial-to-mesenchymal transition (EMT) is a key morphogenetic process that is implicated in the acquisition of stem cell-like properties in different adult tissues, and it is activated in the epicardium after ischemic injury to the heart. We investigated whether EMT is involved in the formation and differentiation of human CSps, revealing that an up-regulation of the expression of EMT-related genes accompanies CSps formation that is relative to primary explant-derived cells and CSp-derived cells grown in a monolayer. EMT and CSps formation is enhanced in the presence of transforming growth factor β1 (TGFβ1) and drastically blocked by the type I TGFβ-receptor inhibitor SB431452, indicating that TGFβ-dependent EMT is essential for the formation of these niche-like 3D-multicellular clusters. Since TGFβ is activated in the myocardium in response to injury, our data suggest that CSps formation mimics an adaptive mechanism that could potentially be enhanced to increase in vivo or ex vivo regenerative potential of adult CPCs.
Collapse
Affiliation(s)
- Elvira Forte
- Department of Molecular Medicine, Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Interstitial fluid flow and cyclic strain differentially regulate cardiac fibroblast activation via AT1R and TGF-β1. Exp Cell Res 2011; 318:75-84. [PMID: 22020089 DOI: 10.1016/j.yexcr.2011.10.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2011] [Revised: 09/30/2011] [Accepted: 10/06/2011] [Indexed: 01/31/2023]
Abstract
Cardiac fibroblasts are exposed to both cyclic strain and interstitial fluid flow in the myocardium. The balance of these stimuli is affected by fibrotic scarring, during which the fibroblasts transition to a myofibroblast phenotype. The present study investigates the mechanisms by which cardiac fibroblasts seeded in three-dimensional (3D) collagen gels differentiate between strain and fluid flow. Neonatal cardiac fibroblast-seeded 3D collagen gels were exposed to interstitial flow and/or cyclic strain and message levels of collagens type I and III, transforming growth factor β1 (TGF-β1), and α-smooth muscle actin (α-SMA) were assessed. Flow was found to significantly increase and strain to decrease expression of myofibroblast markers. Corresponding immunofluorescence indicated that flow and strain differentially regulated α-SMA protein expression. The effect of flow was inhibited by exposure to losartan, an angiotensin II type 1 receptor (AT1R) blocker, and by introduction of shRNA constructs limiting AT1R expression. Blocking of TGF-β also inhibited the myofibroblast transition, suggesting that flow-mediated cell signaling involved both AT1R and TGF-β1. Reduced smad2 phosphorylation in response to cyclic strain suggested that TGF-β is part of the mechanism by which cardiac fibroblasts differentiate between strain-induced and flow-induced mechanical stress. Our experiments show that fluid flow and mechanical deformation have distinct effects on cardiac fibroblast phenotype. Our data suggest a mechanism in which fluid flow directly acts on AT1R and causes increased TGF-β1 expression, whereas cyclic strain reduces activation of smad proteins. These results have relevance to the pathogenesis and treatment of heart failure.
Collapse
|