1
|
Cliff CL, Squires PE, Hills CE. Tonabersat suppresses priming/activation of the NOD-like receptor protein-3 (NLRP3) inflammasome and decreases renal tubular epithelial-to-macrophage crosstalk in a model of diabetic kidney disease. Cell Commun Signal 2024; 22:351. [PMID: 38970061 PMCID: PMC11225428 DOI: 10.1186/s12964-024-01728-1] [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: 03/15/2024] [Accepted: 06/26/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Accompanied by activation of the NOD-like receptor protein 3 (NLRP3) inflammasome, aberrant connexin 43 (Cx43) hemichannel-mediated ATP release is situated upstream of inflammasome assembly and inflammation and contributes to multiple secondary complications of diabetes and associated cardiometabolic comorbidities. Evidence suggests there may be a link between Cx43 hemichannel activity and inflammation in the diabetic kidney. The consequences of blocking tubular Cx43 hemichannel-mediated ATP release in priming/activation of the NLRP3 inflammasome in a model of diabetic kidney disease (DKD) was investigated. We examined downstream markers of inflammation and the proinflammatory and chemoattractant role of the tubular secretome on macrophage recruitment and activation. METHODS Analysis of human transcriptomic data from the Nephroseq repository correlated gene expression to renal function in DKD. Primary human renal proximal tubule epithelial cells (RPTECs) and monocyte-derived macrophages (MDMs) were cultured in high glucose and inflammatory cytokines as a model of DKD to assess Cx43 hemichannel activity, NLRP3 inflammasome activation and epithelial-to-macrophage paracrine-mediated crosstalk. Tonabersat assessed a role for Cx43 hemichannels. RESULTS Transcriptomic analysis from renal biopsies of patients with DKD showed that increased Cx43 and NLRP3 expression correlated with declining glomerular filtration rate (GFR) and increased proteinuria. In vitro, Tonabersat blocked glucose/cytokine-dependant increases in Cx43 hemichannel-mediated ATP release and reduced expression of inflammatory markers and NLRP3 inflammasome activation in RPTECs. We observed a reciprocal relationship in which NLRP3 activity exacerbated increased Cx43 expression and hemichannel-mediated ATP release, events driven by nuclear factor kappa-B (NFκB)-mediated priming and Cx43 hemichannel opening, changes blocked by Tonabersat. Conditioned media (CM) from RPTECs treated with high glucose/cytokines increased expression of inflammatory markers in MDMs, an effect reduced when macrophages were pre-treated with Tonabersat. Co-culture using conditioned media from Tonabersat-treated RPTECs dampened macrophage inflammatory marker expression and reduced macrophage migration. CONCLUSION Using a model of DKD, we report for the first time that high glucose and inflammatory cytokines trigger aberrant Cx43 hemichannel activity, events that instigate NLRP3-induced inflammation in RPTECs and epithelial-to-macrophage crosstalk. Recapitulating observations previously reported in diabetic retinopathy, these data suggest that Cx43 hemichannel blockers (i.e., Tonabersat) may dampen multi-system damage observed in secondary complications of diabetes.
Collapse
Affiliation(s)
- C L Cliff
- Joseph Banks Laboratories, School of Life and Environmental Sciences, University of Lincoln, Lincoln, LN6 7DL, UK
| | - P E Squires
- Joseph Banks Laboratories, School of Life and Environmental Sciences, University of Lincoln, Lincoln, LN6 7DL, UK
| | - C E Hills
- Joseph Banks Laboratories, School of Life and Environmental Sciences, University of Lincoln, Lincoln, LN6 7DL, UK.
| |
Collapse
|
2
|
Lucero CM, Navarro L, Barros-Osorio C, Cáceres-Conejeros P, Orellana JA, Gómez GI. Activation of Pannexin-1 channels causes cell dysfunction and damage in mesangial cells derived from angiotensin II-exposed mice. Front Cell Dev Biol 2024; 12:1387234. [PMID: 38660621 PMCID: PMC11041381 DOI: 10.3389/fcell.2024.1387234] [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: 02/17/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Chronic kidney disease (CKD) is a prevalent health concern associated with various pathological conditions, including hypertensive nephropathy. Mesangial cells are crucial in maintaining glomerular function, yet their involvement in CKD pathogenesis remains poorly understood. Recent evidence indicates that overactivation of Pannexin-1 (Panx1) channels could contribute to the pathogenesis and progression of various diseases. Although Panx1 is expressed in the kidney, its contribution to the dysfunction of renal cells during pathological conditions remains to be elucidated. This study aimed to investigate the impact of Panx1 channels on mesangial cell function in the context of hypertensive nephropathy. Using an Ang II-infused mouse model and primary mesangial cell cultures, we demonstrated that in vivo exposure to Ang II sensitizes cultured mesangial cells to show increased alterations when they are subjected to subsequent in vitro exposure to Ang II. Particularly, mesangial cell cultures treated with Ang II showed elevated activity of Panx1 channels and increased release of ATP. The latter was associated with enhanced basal intracellular Ca2+ ([Ca2+]i) and increased ATP-mediated [Ca2+]i responses. These effects were accompanied by increased lipid peroxidation and reduced cell viability. Crucially, all the adverse impacts evoked by Ang II were prevented by the blockade of Panx1 channels, underscoring their critical role in mediating cellular dysfunction in mesangial cells. By elucidating the mechanisms by which Ang II negatively impacts mesangial cell function, this study provides valuable insights into the pathogenesis of renal damage in hypertensive nephropathy.
Collapse
Affiliation(s)
- Claudia M. Lucero
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Laura Navarro
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Cristián Barros-Osorio
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Patricio Cáceres-Conejeros
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| | - Juan A. Orellana
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gonzalo I. Gómez
- Institute of Biomedical Sciences, Faculty of Health Sciences, Universidad Autónoma de Chile, Santiago, Chile
| |
Collapse
|
3
|
Kowalewska PM, Milkovich SL, Goldman D, Sandow SL, Ellis CG, Welsh DG. Capillary oxygen regulates demand-supply coupling by triggering connexin40-mediated conduction: Rethinking the metabolic hypothesis. Proc Natl Acad Sci U S A 2024; 121:e2303119121. [PMID: 38349880 PMCID: PMC10895355 DOI: 10.1073/pnas.2303119121] [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: 02/26/2023] [Accepted: 12/21/2023] [Indexed: 02/15/2024] Open
Abstract
Coupling red blood cell (RBC) supply to O2 demand is an intricate process requiring O2 sensing, generation of a stimulus, and signal transduction that alters upstream arteriolar tone. Although actively debated, this process has been theorized to be induced by hypoxia and to involve activation of endothelial inwardly rectifying K+ channels (KIR) 2.1 by elevated extracellular K+ to trigger conducted hyperpolarization via connexin40 (Cx40) gap junctions to upstream resistors. This concept was tested in resting healthy skeletal muscle of Cx40-/- and endothelial KIR2.1-/- mice using state-of-the-art live animal imaging where the local tissue O2 environment was manipulated using a custom gas chamber. Second-by-second capillary RBC flow responses were recorded as O2 was altered. A stepwise drop in PO2 at the muscle surface increased RBC supply in capillaries of control animals while elevated O2 elicited the opposite response; capillaries were confirmed to express Cx40. The RBC flow responses were rapid and tightly coupled to O2; computer simulations did not support hypoxia as a driving factor. In contrast, RBC flow responses were significantly diminished in Cx40-/- mice. Endothelial KIR2.1-/- mice, on the other hand, reacted normally to O2 changes, even when the O2 challenge was targeted to a smaller area of tissue with fewer capillaries. Conclusively, microvascular O2 responses depend on coordinated electrical signaling via Cx40 gap junctions, and endothelial KIR2.1 channels do not initiate the event. These findings reconceptualize the paradigm of blood flow regulation in skeletal muscle and how O2 triggers this process in capillaries independent of extracellular K+.
Collapse
Affiliation(s)
- Paulina M. Kowalewska
- Robarts Research Institute, University of Western Ontario, London, ONN6A 5B7, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, ONN6A 5B7, Canada
| | | | - Daniel Goldman
- Department of Medical Biophysics, University of Western Ontario, London, ONN6A 5B7, Canada
| | - Shaun L. Sandow
- School of Health, University of the Sunshine Coast, Maroochydore, QLD4556, Australia
- School of Clinical Medicine, University of Queensland, St. Lucia, QLD4072, Australia
| | - Christopher G. Ellis
- Robarts Research Institute, University of Western Ontario, London, ONN6A 5B7, Canada
- Department of Medical Biophysics, University of Western Ontario, London, ONN6A 5B7, Canada
| | - Donald G. Welsh
- Robarts Research Institute, University of Western Ontario, London, ONN6A 5B7, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, ONN6A 5B7, Canada
| |
Collapse
|
4
|
Roger E, Chadjichristos CE, Kavvadas P, Price GW, Cliff CL, Hadjadj S, Renciot J, Squires PE, Hills CE. Connexin-43 hemichannels orchestrate NOD-like receptor protein-3 (NLRP3) inflammasome activation and sterile inflammation in tubular injury. Cell Commun Signal 2023; 21:263. [PMID: 37770948 PMCID: PMC10536814 DOI: 10.1186/s12964-023-01245-7] [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: 05/26/2023] [Accepted: 07/23/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Without a viable cure, chronic kidney disease is a global health concern. Inflammatory damage in and around the renal tubules dictates disease severity and is contributed to by multiple cell types. Activated in response to danger associated molecular patterns (DAMPs) including ATP, the NOD-like receptor protein-3 (NLRP3) inflammasome is integral to this inflammation. In vivo, we have previously observed that increased expression of Connexin 43 (Cx43) is linked to inflammation in chronic kidney disease (CKD) whilst in vitro studies in human proximal tubule cells highlight a role for aberrant Cx43 hemichannel mediated ATP release in tubule injury. A role for Cx43 hemichannels in priming and activation of the NLRP3 inflammasome in tubule epithelial cells remains to be determined. METHODS Using the Nephroseq database, analysis of unpublished transcriptomic data, examined gene expression and correlation in human CKD. The unilateral ureteral obstruction (UUO) mouse model was combined with genetic (tubule-specific Cx43 knockout) and specific pharmacological blockade of Cx43 (Peptide5), to explore a role for Cx43-hemichannels in tubule damage. Human primary tubule epithelial cells were used as an in vitro model of CKD. RESULTS Increased Cx43 and NLRP3 expression correlates with declining glomerular filtration rate and increased proteinuria in biopsies isolated from patients with CKD. Connexin 43-tubule deletion prior to UUO protected against tubular injury, increased expression of proinflammatory molecules, and significantly reduced NLRP3 expression and downstream signalling mediators. Accompanied by a reduction in F4/80 macrophages and fibroblast specific protein (FSP1+) fibroblasts, Cx43 specific hemichannel blocker Peptide5 conferred similar protection in UUO mice. In vitro, Peptide5 determined that increased Cx43-hemichannel activity primes and activates the NLRP3 inflammasome via ATP-P2X7 receptor signalling culminating in increased secretion of chemokines and cytokines, each of which are elevated in individuals with CKD. Inhibition of NLRP3 and caspase 1 similarly decreased markers of tubular injury, whilst preventing the perpetual increase in Cx43-hemichannel activity. CONCLUSION Aberrant Cx43-hemichannel activity in kidney tubule cells contributes to tubule inflammation via activation of the NLRP3 inflammasome and downstream paracrine mediated cell signalling. Use of hemichannel blockers in targeting Cx43-hemichannels is an attractive future therapeutic target to slow or prevent disease progression in CKD. Video Abstract.
Collapse
Affiliation(s)
- Elena Roger
- Batiment Recherche, INSERM, UMR-S1155, Tenon Hospital, 4 Rue de la Chine, Paris, 75020, France
- Faculty of Medicine, Sorbonne University, Paris, 75013, France
| | - Christos E Chadjichristos
- Batiment Recherche, INSERM, UMR-S1155, Tenon Hospital, 4 Rue de la Chine, Paris, 75020, France
- Faculty of Medicine, Sorbonne University, Paris, 75013, France
| | - Panagiotis Kavvadas
- Batiment Recherche, INSERM, UMR-S1155, Tenon Hospital, 4 Rue de la Chine, Paris, 75020, France
- Faculty of Medicine, Sorbonne University, Paris, 75013, France
| | - Gareth W Price
- Joseph Banks Laboratories, School of Life and Environmental Sciences, University of Lincoln, Lincoln, LN6 7DL, UK
| | - Chelsy L Cliff
- Joseph Banks Laboratories, School of Life and Environmental Sciences, University of Lincoln, Lincoln, LN6 7DL, UK
| | - Safia Hadjadj
- Batiment Recherche, INSERM, UMR-S1155, Tenon Hospital, 4 Rue de la Chine, Paris, 75020, France
- Faculty of Medicine, Sorbonne University, Paris, 75013, France
| | - Jessy Renciot
- Batiment Recherche, INSERM, UMR-S1155, Tenon Hospital, 4 Rue de la Chine, Paris, 75020, France
- Faculty of Medicine, Sorbonne University, Paris, 75013, France
| | - Paul E Squires
- Joseph Banks Laboratories, School of Life and Environmental Sciences, University of Lincoln, Lincoln, LN6 7DL, UK
| | - Claire E Hills
- Joseph Banks Laboratories, School of Life and Environmental Sciences, University of Lincoln, Lincoln, LN6 7DL, UK.
| |
Collapse
|
5
|
Dai L, Yu W, Yu Y. New strategy of using double-network hydrogel extravascular stent for preventing venous graft restenosis after coronary artery bypass grafting. Perfusion 2023; 38:1240-1249. [PMID: 35511059 DOI: 10.1177/02676591221099813] [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] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Effective therapies for the prevention of vein graft failure, which frequently occurs in coronary artery bypass grafting (CABG) due to intimal hyperplasia (IH), are still lacking. Here, we investigated the effects of the perivenous application of double-network hydrogel on vein grafts in carotid artery bypass grafting in a rabbit model. METHODS Healthy New Zealand white rabbits were randomized into the following groups: no graft, graft, or graft + Double-network hydrogel external stent (DNHES). The rabbits' carotid artery was bypassed via the jugular vein. Double-network hydrogel external stent was wrapped around the jugular graft after the anastomoses were completed. Blood flow parameters and tissue histology of the vein grafts were evaluated. RESULTS Compared with the untreated vein grafts at 12 weeks after the surgery, the DNHES significantly improved graft flow, attenuated intimal and medial thickening, reduced the anti-proliferating cell nuclear antigen proliferation index of the vein grafts, decreased the mRNA and protein expression of Mitogen-Activated Protein Kinase (MAPK) and Transforming Growth Factor-β (TGF-β), and increased the mRNA and protein expression of endothelial Nitric Oxide Synthase (eNOS). CONCLUSION The perivenous application of DNHES exerts beneficial effects on vein grafts, reduces the inflammatory response in carotid artery bypass grafting in a rabbit model, and appears to be a safe and promising strategy to prevent vein graft failure.
Collapse
Affiliation(s)
- Longsheng Dai
- Department of Cardiac Surgery, Beijing An Zhen Hospital, Capital Medical University, Beijing, China
| | - Wenyuan Yu
- Department of Cardiac Surgery, Beijing An Zhen Hospital, Capital Medical University, Beijing, China
| | - Yang Yu
- Department of Cardiac Surgery, Beijing An Zhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Kiełbowski K, Bakinowska E, Pawlik A. The Potential Role of Connexins in the Pathogenesis of Atherosclerosis. Int J Mol Sci 2023; 24:ijms24032600. [PMID: 36768920 PMCID: PMC9916887 DOI: 10.3390/ijms24032600] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/29/2022] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
Connexins (Cx) are members of a protein family which enable extracellular and intercellular communication through hemichannels and gap junctions (GJ), respectively. Cx take part in transporting important cell-cell messengers such as 3',5'-cyclic adenosine monophosphate (cAMP), adenosine triphosphate (ATP), and inositol 1,4,5-trisphosphate (IP3), among others. Therefore, they play a significant role in regulating cell homeostasis, proliferation, and differentiation. Alterations in Cx distribution, degradation, and post-translational modifications have been correlated with cancers, as well as cardiovascular and neurological diseases. Depending on the isoform, Cx have been shown either to promote or suppress the development of atherosclerosis, a progressive inflammatory disease affecting large and medium-sized arteries. Cx might contribute to the progression of the disease by enhancing endothelial dysfunction, monocyte recruitment, vascular smooth muscle cell (VSMC) activation, or by inhibiting VSMC autophagy. Inhibition or modulation of the expression of specific isoforms could suppress atherosclerotic plaque formation and diminish pro-inflammatory conditions. A better understanding of the complexity of atherosclerosis pathophysiology linked with Cx could result in developing novel therapeutic strategies. This review aims to present the role of Cx in the pathogenesis of atherosclerosis and discusses whether they can become novel therapeutic targets.
Collapse
|
7
|
Tang ACW, Chen CK, Wu SY, Tang SFT. Improvement of Pain and Function by Using Botulinum Toxin Type A Injection in Patients with an Osteoarthritic Knee with Patellar Malalignment: An Electromyographic Study. Life (Basel) 2022; 13:life13010095. [PMID: 36676044 PMCID: PMC9863177 DOI: 10.3390/life13010095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/24/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
Objective: To determine the pain and electromyographic (EMG) amplitude ratio of the vastus medialis oblique (VMO) to the vastus lateralis (VL) after botulinum toxin type A (BTA) was injected in the bilateral osteoarthritic knee of patients with patellar malalignment for analysis. Material and methods: A total of fifteen patients were recruited; the more symptomatic knee of each patient received a BTA injection (BTA side). The other set of patients were left untreated. In all, fifteen healthy participants comprised the control group. The Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) and numeric rating scale (NRS) for pain were assessed. The EMG amplitude of VL and VMO activity was recorded using an isokinetic dynamometer and synchronized using the BIOPAC MP100. The data were collected before and at 4, 8, and 12 weeks post−BTA injection. Results: The EMG ratios of the patient group were lower than those of the control group at all testing velocities (p < 0.05). The VMO/VL ratio improved significantly on the BTA side only. The VMO/VL ratios on the BTA side were higher than those on the untreated side (p < 0.05). Knee pain decreased significantly after the BTA injection. The EMG ratios were negatively correlated with the NRS and WOMAC scores. Conclusion: BTA injection effectively reduces knee pain and restores the EMG ratio between the VMO and VL.
Collapse
Affiliation(s)
- Alice Chu Wen Tang
- Department of Physical Medicine and Rehabilitation, Fu Jen Catholic University Hospital, New Taipei City 234, Taiwan
| | - Chih-Kuang Chen
- Department of Physical Medicine and Rehabilitation, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333423, Taiwan
| | - Szu Yuan Wu
- Division of Radiation Oncology and Big Data Center, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan 265501, Taiwan
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, Taipei 24205, Taiwan
- Centers for Regional Anesthesia and Pain Medicine, Taipei Municipal Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Simon F. T. Tang
- Department of Physical Medicine and Rehabilitation, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333423, Taiwan
- Department of Physical Medicine and Rehabilitation, Lotung Poh-Ai Hospital, Yilan 265501, Taiwan
- Correspondence: ; Tel.: +886-3-9543131 (ext. 3300); Fax: +886-3-9565042
| |
Collapse
|
8
|
Hypertensive Nephropathy: Unveiling the Possible Involvement of Hemichannels and Pannexons. Int J Mol Sci 2022; 23:ijms232415936. [PMID: 36555574 PMCID: PMC9785367 DOI: 10.3390/ijms232415936] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Hypertension is one of the most common risk factors for developing chronic cardiovascular diseases, including hypertensive nephropathy. Within the glomerulus, hypertension causes damage and activation of mesangial cells (MCs), eliciting the production of large amounts of vasoactive and proinflammatory agents. Accordingly, the activation of AT1 receptors by the vasoactive molecule angiotensin II (AngII) contributes to the pathogenesis of renal damage, which is mediated mostly by the dysfunction of intracellular Ca2+ ([Ca2+]i) signaling. Similarly, inflammation entails complex processes, where [Ca2+]i also play crucial roles. Deregulation of this second messenger increases cell damage and promotes fibrosis, reduces renal blood flow, and impairs the glomerular filtration barrier. In vertebrates, [Ca2+]i signaling depends, in part, on the activity of two families of large-pore channels: hemichannels and pannexons. Interestingly, the opening of these channels depends on [Ca2+]i signaling. In this review, we propose that the opening of channels formed by connexins and/or pannexins mediated by AngII induces the ATP release to the extracellular media, with the subsequent activation of purinergic receptors. This process could elicit Ca2+ overload and constitute a feed-forward mechanism, leading to kidney damage.
Collapse
|
9
|
Sedovy MW, Leng X, Leaf MR, Iqbal F, Payne LB, Chappell JC, Johnstone SR. Connexin 43 across the Vasculature: Gap Junctions and Beyond. J Vasc Res 2022; 60:101-113. [PMID: 36513042 PMCID: PMC11073551 DOI: 10.1159/000527469] [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: 05/25/2022] [Accepted: 09/26/2022] [Indexed: 12/15/2022] Open
Abstract
Connexin 43 (Cx43) is essential to the function of the vasculature. Cx43 proteins form gap junctions that allow for the exchange of ions and molecules between vascular cells to facilitate cell-to-cell signaling and coordinate vasomotor activity. Cx43 also has intracellular signaling functions that influence vascular cell proliferation and migration. Cx43 is expressed in all vascular cell types, although its expression and function vary by vessel size and location. This includes expression in vascular smooth muscle cells (vSMC), endothelial cells (EC), and pericytes. Cx43 is thought to coordinate homocellular signaling within EC and vSMC. Cx43 gap junctions also function as conduits between different cell types (heterocellular signaling), between EC and vSMC at the myoendothelial junction, and between pericyte and EC in capillaries. Alterations in Cx43 expression, localization, and post-translational modification have been identified in vascular disease states, including atherosclerosis, hypertension, and diabetes. In this review, we discuss the current understanding of Cx43 localization and function in healthy and diseased blood vessels across all vascular beds.
Collapse
Affiliation(s)
- Meghan W. Sedovy
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
- Translational Biology, Medicine, And Health Graduate Program, Virginia Tech, Blacksburg, VA, USA
| | - Xinyan Leng
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
| | - Melissa R. Leaf
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| | - Farwah Iqbal
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| | - Laura Beth Payne
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
| | - John C. Chappell
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
| | - Scott R. Johnstone
- The Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, 4 Riverside Circle, Roanoke, VA, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|
10
|
Ren KW, Yu XH, Gu YH, Xie X, Wang Y, Wang SH, Li HH, Bi HL. Cardiac-specific knockdown of Bhlhe40 attenuates angiotensin II (Ang II)-Induced atrial fibrillation in mice. Front Cardiovasc Med 2022; 9:957903. [PMID: 36304536 PMCID: PMC9592817 DOI: 10.3389/fcvm.2022.957903] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Atrial fibrosis and atrial inflammation are associated with the pathogenesis of atrial fibrillation (AF). Basic helix–loop–helix family member E40 (Bhlhe40) is an important transcription factor, which is involved in tumors, inflammation, apoptosis, viral infection, and hypoxia. However, its role and molecular mechanism in AF remain unclear. In this study, a mouse model of AF was induced by Ang II infusion. The atrial diameter was evaluated using echocardiography. Induction and duration of AF were measured by programmed electrical stimulation. Atrial structural remodeling was detected using routine histologic examinations. Our results showed that Bhlhe40 was significantly upregulated in angiotensin II (Ang II)-stimulated atrial cardiomyocytes and atrial tissues and in tissues from patients with AF. Cardiac-specific knockdown of Bhlhe40 in mice by a type 9 recombinant adeno-associated virus (rAAV9)-shBhlhe40 significantly ameliorated Ang II-induced atrial dilatation, atrial fibrosis, and atrial inflammation, as well as the inducibility and duration of AF. Mechanistically, cardiac-specific knockdown of Bhlhe40 attenuated Ang II-induced activation of NF-κB/NLRP3, TGF-1β/Smad2 signals, the increased expression of CX43, and the decreased expression of Kv4.3 in the atria. This is the first study to suggest that Bhlhe40 is a novel regulator of AF progression, and identifying Bhlhe40 may be a new therapeutic target for hypertrophic remodeling and heart failure.
Collapse
Affiliation(s)
- Kai-Wen Ren
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiao-Hong Yu
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yu-Hui Gu
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xin Xie
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yu Wang
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shi-hao Wang
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hui-Hua Li
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China,*Correspondence: Hui-Hua Li,
| | - Hai-Lian Bi
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China,Hai-Lian Bi,
| |
Collapse
|
11
|
Nguyen TLL, Jin Y, Kim L, Heo KS. Inhibitory effects of 6'-sialyllactose on angiotensin II-induced proliferation, migration, and osteogenic switching in vascular smooth muscle cells. Arch Pharm Res 2022; 45:658-670. [PMID: 36070173 DOI: 10.1007/s12272-022-01404-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 08/25/2022] [Indexed: 12/14/2022]
Abstract
Excessive production and migration of vascular smooth muscle cells (VSMCs) are associated with vascular remodeling that causes vascular diseases, such as restenosis and hypertension. Angiotensin II (Ang II) stimulation is a key factor in inducing abnormal VSMC function. This study aimed to investigate the effects of 6'-sialyllactose (6'SL), a human milk oligosaccharide, on Ang II-stimulated cell proliferation, migration and osteogenic switching in rat aortic smooth muscle cells (RASMCs) and human aortic smooth muscle cells (HASMCs). Compared with the control group, Ang II increased cell proliferation by activating MAPKs, including ERK1/2/p90RSK/Akt/mTOR and JNK pathways. However, 6'SL reversed Ang II-stimulated cell proliferation and the ERK1/2/p90RSK/Akt/mTOR pathways in RASMCs and HASMCs. Moreover, 6'SL suppressed Ang II-stimulated cell cycle progression from G0/G1 to S and G2/M phases in RASMCs. Furthermore, 6'SL effectively inhibited cell migration by downregulating NF-κB-mediated MMP2/9 and VCAM-1 expression levels. Interestingly, in RASMCs, 6'SL attenuated Ang II-induced osteogenic switching by reducing the production of p90RSK-mediated c-fos and JNK-mediated c-jun, leading to the downregulation of AP-1-mediated osteopontin production. Taken together, our data suggest that 6'SL inhibits Ang II-induced VSMC proliferation and migration by abolishing the ERK1/2/p90RSK-mediated Akt and NF-κB signaling pathways, respectively, and osteogenic switching by suppressing p90RSK- and JNK-mediated AP-1 activity.
Collapse
Affiliation(s)
- Thuy Le Lam Nguyen
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon, South Korea
| | - Yujin Jin
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon, South Korea
| | - Lila Kim
- GeneChem Inc., Daejeon, South Korea
| | - Kyung-Sun Heo
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, Daejeon, South Korea.
| |
Collapse
|
12
|
TNF-α Plus IL-1β Induces Opposite Regulation of Cx43 Hemichannels and Gap Junctions in Mesangial Cells through a RhoA/ROCK-Dependent Pathway. Int J Mol Sci 2022; 23:ijms231710097. [PMID: 36077498 PMCID: PMC9456118 DOI: 10.3390/ijms231710097] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
Connexin 43 (Cx43) is expressed in kidney tissue where it forms hemichannels and gap junction channels. However, the possible functional relationship between these membrane channels and their role in damaged renal cells remains unknown. Here, analysis of ethidium uptake and thiobarbituric acid reactive species revealed that treatment with TNF-α plus IL-1β increases Cx43 hemichannel activity and oxidative stress in MES-13 cells (a cell line derived from mesangial cells), and in primary mesangial cells. The latter was also accompanied by a reduction in gap junctional communication, whereas Western blotting assays showed a progressive increase in phosphorylated MYPT (a target of RhoA/ROCK) and Cx43 upon TNF-α/IL-1β treatment. Additionally, inhibition of RhoA/ROCK strongly antagonized the TNF-α/IL-1β-induced activation of Cx43 hemichannels and reduction in gap junctional coupling. We propose that activation of Cx43 hemichannels and inhibition of cell-cell coupling during pro-inflammatory conditions could contribute to oxidative stress and damage of mesangial cells via the RhoA/ROCK pathway.
Collapse
|
13
|
Zhou Y, Suo W, Zhang X, Lv J, Liu Z, Liu R. Roles and mechanisms of quercetin on cardiac arrhythmia: A review. Biomed Pharmacother 2022; 153:113447. [DOI: 10.1016/j.biopha.2022.113447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 11/02/2022] Open
|
14
|
Reversing Cardiac Hypertrophy at the Source Using a Cardiac Targeting Peptide Linked to miRNA106a: Targeting Genes That Cause Cardiac Hypertrophy. Pharmaceuticals (Basel) 2022; 15:ph15070871. [PMID: 35890169 PMCID: PMC9317130 DOI: 10.3390/ph15070871] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/06/2022] [Accepted: 07/09/2022] [Indexed: 02/04/2023] Open
Abstract
Causes and treatments for heart failure (HF) have been investigated for over a century culminating in data that have led to numerous pharmacological and surgical therapies. Unfortunately, to date, even with the most current treatments, HF remains a progressive disease with no therapies targeting the cardiomyocytes directly. Technological advances within the past two to three years have brought about new paradigms for treating many diseases that previously had been extremely difficult to resolve. One of these new paradigms has been a shift from pharmacological agents to antisense technology (e.g., microRNAs) to target the molecular underpinnings of pathological processes leading to disease onset. Although this paradigm shift may have been postulated over a decade ago, only within the past few years has it become feasible. Here, we show that miRNA106a targets genes that, when misregulated, have been shown to cause hypertrophy and eventual HF. The addition of miRNA106a suppresses misexpressed HF genes and reverses hypertrophy. Most importantly, using a cardiac targeting peptide reversibly linked to miRNA106a, we show delivery is specific to cardiomyocytes.
Collapse
|
15
|
Qin X, Gao A, Hou X, Xu X, Chen L, Sun L, Hao Y, Shi Y. Connexins may play a critical role in cigarette smoke-induced pulmonary hypertension. Arch Toxicol 2022; 96:1609-1621. [PMID: 35344070 DOI: 10.1007/s00204-022-03274-6] [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: 02/17/2022] [Accepted: 03/02/2022] [Indexed: 11/02/2022]
Abstract
Pulmonary hypertension (PH) is a chronic progressive disease characterized by pulmonary vasoconstriction and remodeling. It causes a gradual increase in pulmonary vascular resistance leading to right-sided heart failure, and may be fatal. Chronic exposure to cigarette smoke (CS) is an essential risk factor for PH group 3; however, smoking continues to be prevalent and smoking cessation is reported to be difficult. A majority of smokers exhibit PH, which leads to a concomitant increase in the risk of mortality. The current treatments for PH group 3 focus on vasodilation and long-term oxygen supplementation, and fail to stop or reverse PH-associated continuous vascular remodeling. Recent studies have suggested that pulmonary vascular endothelial dysfunction induced by CS exposure may be an initial event in the natural history of PH, which in turn may be associated with abnormal alterations in connexin (Cx) expression. The relationship between Cx and CS-induced PH development has not yet been directly investigated. Therefore, this review will describe the roles of CS and Cx in the development of PH and discuss the related downstream pathways. We also discuss the possible role of Cx in CS-induced PH. It is hoped that this review may provide new perspectives for early intervention.
Collapse
Affiliation(s)
- Xiaojiang Qin
- School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China.
- China Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China.
| | - Anqi Gao
- School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Xiaomin Hou
- Department of Pharmacology, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
- China Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Xinrong Xu
- School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Liangjin Chen
- School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Lin Sun
- School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Yuxuan Hao
- School of Public Health, Shanxi Medical University, 56 Xinjian South Road, Taiyuan, 030001, Shanxi, China
| | - Yiwei Shi
- Department of Respiratory and Critical Care Medicine, Shanxi Medical University Affiliated First Hospital, 85 Jiefang South Road, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
16
|
Haefliger JA, Meda P, Alonso F. Endothelial Connexins in Developmental and Pathological Angiogenesis. Cold Spring Harb Perspect Med 2022; 12:a041158. [PMID: 35074793 PMCID: PMC9159259 DOI: 10.1101/cshperspect.a041158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Connexins (Cxs) constitute a large family of transmembrane proteins that form gap junction channels, which enable the direct transfer of small signaling molecules from cell to cell. In blood vessels, Cx channels allow the endothelial cells (ECs) to respond to external and internal cues as a whole and, thus, contribute to the maintenance of vascular homeostasis. While the role of Cxs has been extensively studied in large arteries, a growing body of evidence suggests that they also play a role in the formation of microvascular networks. Since the formation of new blood vessels requires the coordinated response of ECs to external stimuli, endothelial Cxs may play an important role there. Recent studies in developmental and pathologic models reveal that EC Cxs regulate physiological and pathological angiogenesis through canonical and noncanonical functions, making these proteins potential therapeutic targets for the development of new strategies aimed at a better control of angiogenesis.
Collapse
Affiliation(s)
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva, Medical Center, 1211 Geneva, Switzerland
| | - Florian Alonso
- Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), Université de Bordeaux, 33076 Bordeaux, France
| |
Collapse
|
17
|
Sathiyanadan K, Alonso F, Domingos-Pereira S, Santoro T, Hamard L, Cesson V, Meda P, Nardelli-Haefliger D, Haefliger JA. Targeting Endothelial Connexin37 Reduces Angiogenesis and Decreases Tumor Growth. Int J Mol Sci 2022; 23:2930. [PMID: 35328350 PMCID: PMC8948817 DOI: 10.3390/ijms23062930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 12/13/2022] Open
Abstract
Connexin37 (Cx37) and Cx40 form intercellular channels between endothelial cells (EC), which contribute to the regulation of the functions of vessels. We previously documented the participation of both Cx in developmental angiogenesis and have further shown that loss of Cx40 decreases the growth of different tumors. Here, we report that loss of Cx37 reduces (1) the in vitro proliferation of primary human EC; (2) the vascularization of subcutaneously implanted matrigel plugs in Cx37-/- mice or in WT using matrigel plugs supplemented with a peptide targeting Cx37 channels; (3) tumor angiogenesis; and (4) the growth of TC-1 and B16 tumors, resulting in a longer mice survival. We further document that Cx37 and Cx40 function in a collaborative manner to promote tumor growth, inasmuch as the injection of a peptide targeting Cx40 into Cx37-/- mice decreased the growth of TC-1 tumors to a larger extent than after loss of Cx37. This loss did not alter vessel perfusion, mural cells coverage and tumor hypoxia compared to tumors grown in WT mice. The data show that Cx37 is relevant for the control of EC proliferation and growth in different tumor models, suggesting that it may be a target, alone or in combination with Cx40, in the development of anti-tumoral treatments.
Collapse
Affiliation(s)
- Karthik Sathiyanadan
- Department of Urology, Lausanne University Hospital, 1011 Lausanne, Switzerland; (K.S.); (S.D.-P.); (V.C.); (D.N.-H.)
| | - Florian Alonso
- Laboratory for the Bioengineering of Tissues (BioTis-INSERM U1026), Université de Bordeaux, 33607 Bordeaux, France;
| | - Sonia Domingos-Pereira
- Department of Urology, Lausanne University Hospital, 1011 Lausanne, Switzerland; (K.S.); (S.D.-P.); (V.C.); (D.N.-H.)
| | - Tania Santoro
- Department of Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland; (T.S.); (L.H.)
| | - Lauriane Hamard
- Department of Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland; (T.S.); (L.H.)
| | - Valérie Cesson
- Department of Urology, Lausanne University Hospital, 1011 Lausanne, Switzerland; (K.S.); (S.D.-P.); (V.C.); (D.N.-H.)
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, Medical Center, University of Geneva, 1206 Geneva, Switzerland;
| | - Denise Nardelli-Haefliger
- Department of Urology, Lausanne University Hospital, 1011 Lausanne, Switzerland; (K.S.); (S.D.-P.); (V.C.); (D.N.-H.)
| | | |
Collapse
|
18
|
Gao RJ, Zhang AM, Jia QH, Dang ZT, Tian T, Zhang JR, Cao N, Tang XC, Ma KT, Li L, Si JQ. The promoting role of Cx43 on the proliferation and migration of arterial smooth muscle cells for angiotensin II-dependent hypertension. Pulm Pharmacol Ther 2021; 70:102072. [PMID: 34428599 DOI: 10.1016/j.pupt.2021.102072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/03/2021] [Accepted: 08/16/2021] [Indexed: 11/15/2022]
Abstract
BACKGROUND Recent studies have shown that endothelin-1 and angiotensin II (AngII) can increase gap junctional intercellular communication (GJIC) by activating Mitogen-activated protein kinases (MAPKs) pathway. However, not only the precise interaction of AngII with Connexin43(Cx43) and the associated functions remain unclear, but also the regulatory role of Cx43 on the AngII-mediated promotion proliferation and migration of VSMCs is poorly understood. MATERIAL AND METHODS Our research applicated pressure myography measurements, immunofluorescence and Western blot analyses to investigate the changes in physiological indicators in spontaneously hypertensive rats (SHRs) and AngII-stimulated proliferation and migration of A7r5 SMCs(Rat vascular smooth muscle cells). The aim was to elucidate the role of CX43 in hypertension induced by AngII. RESULTS Chronic ramipril (angiotensin converting enzyme inhibitor) management for SHRs significantly attenuated blood pressure and blood vessel wall thickness, also reduced contraction rate in the cerebral artery. The cerebral artery contraction rates, mRNA and protein expression of Cx43, osteopontin (OPN) and proliferating cell nuclear antigen (PCNA) protein expression in the SHR + ramipril and SHR + ramipril + carbenoxolone (CBX, Cx43 specific blocker) groups were significantly lower than those in the SHR group. Cx43 protein expression and Ser368 phosphorylated Cx43 protein levels increased significantly in AngII-stimulated A7r5 cells. However, the levels of phosphorylated Cx43 decreased after pre-treatment with candesartan (AT1 receptor blocker), GF109203X (protein kinase C (PKC) blocker) and U0126 (mitogen-activated protein kinases/extracellular signal-regulated kinase1/2(MEK/ERK1/2)-specific blocker) in AngII-stimulated A7r5 cells. Cx43 was widely distributed in the cell membrane, nucleus, and cytoplasm of the SMCs. Furthermore, pre-treatment of the AngII- stimulated A7r5 cells with Gap26 (Cx43 blocker) significantly inhibited cell migration and decreased the expression levels of MEK1/2, ERK1/2, P-MEK1/2, and P-ERK1/2. CONCLUSION Our research confirms that Cx43 plays an important role in the regulation of proliferation and migration of VSMCs via MEK/ERK and PKC signal pathway in AngII-dependent hypertension.
Collapse
Affiliation(s)
- Rui-Juan Gao
- Department of Physiology, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; Department of Radiology, First Affiliated Hospital of Shihezi University, Shihezi, 832002, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Ai-Mei Zhang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; Department of Cardiology, First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, 832002, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Qi-Hua Jia
- Department of Physiology, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Zi-Ting Dang
- Department of Physiology, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; Department of Commerce, Shanxi Institute of International Trade & Commerce, Xianyang, 712046, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Tian Tian
- Department of Physiology, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Jing-Rong Zhang
- Department of Physiology, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Nan Cao
- Department of Physiology, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Xue-Chun Tang
- Department of Physiology, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Ke-Tao Ma
- Department of Physiology, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China
| | - Li Li
- Department of Physiology, Medical College of Jiaxing University, Jiaxing, 314001, China.
| | - Jun-Qiang Si
- Department of Physiology, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China; Department of Physiology, Huazhong University of Science and Technology of Basic Medical Sciences, Wuhan, 430070, China; NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, 832002, China.
| |
Collapse
|
19
|
Tien (田婷怡) TY, Wu (吳懿哲) YJ, Su (蘇正煌) CH, Wang (王學孝) HH, Hsieh (謝金玲) CL, Wang (王博正) BJ, Su (蘇瑀) Y, Yeh (葉宏一) HI. Reduction of Connexin 43 Attenuates Angiogenic Effects of Human Smooth Muscle Progenitor Cells via Inactivation of Akt and NF-κB Pathway. Arterioscler Thromb Vasc Biol 2021; 41:915-930. [PMID: 33356390 DOI: 10.1161/atvbaha.120.315650] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Circulating progenitor cells possess vasculogenesis property and participate in repair of vascular injury. Cx (connexin) 43-a transmembrane protein constituting gap junctions-is involved in vascular pathology. However, the role of Cx43 in smooth muscle progenitor cells (SPCs) remained unclear. Approach and Results: Human SPCs cultured from CD34+ peripheral blood mononuclear cells expressed smooth muscle cell markers, such as smooth muscle MHC (myosin heavy chain), nonmuscle MHC, calponin, and CD140B, and Cx43 was the most abundant Cx isoform. To evaluate the role of Cx43 in SPCs, short interference RNA was used to knock down Cx43 expression. Cellular activities of SPCs were reduced by Cx43 downregulation. In addition, Cx43 downregulation attenuated angiogenic potential of SPCs in hind limb ischemia mice. Protein array and ELISA of the supernatant from SPCs showed that IL (interleukin)-6, IL-8, and HGF (hepatocyte growth factor) were reduced by Cx43 downregulation. Simultaneously, Cx43 downregulation reduced the phosphorylation of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) and Akt (protein kinase B) pathway and reactivation of NF-κB and Akt using betulinic acid, and SC79 could restore the secretion of growth factors and cytokines. Moreover, FAK (focal adhesion kinase)-Src (proto-oncogene tyrosine-protein kinase Src) activation was increased by Cx43 downregulation, and inactivation of Akt-NF-κB could be restored by Src inhibitor (PP2), indicating that Akt-NF-κB inactivated by Cx43 downregulation arose from FAK-Src activation. Finally, the depressed cellular activities and secretion of SPCs after Cx43 downregulation were restored by FAK inhibitor PF-562271 or PP2. CONCLUSIONS SPCs possess angiogenic potential to repair ischemic tissue mainly through paracrine effects. Gap junction protein Cx43 plays an important role in regulating cellular function and paracrine effects of SPCs through FAK-Src axis.
Collapse
Affiliation(s)
- Ting-Yi Tien (田婷怡)
- Department of Medical Research (T.-Y.T., C.-L.H., B.-J.W.), MacKay Memorial Hospital, Taipei, Taiwan
- Institute of Biopharmaceutical Science/National Yang-Ming University, Taipei, Taiwan (T.-Y.T., Y.S.)
| | - Yih-Jer Wu (吳懿哲)
- Department of Internal Medicine (Y.-J.W., C.-H.S., H.-I.Y.), MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan (Y.-J.W., C.-H.S., H.-H.W.)
| | - Cheng-Huang Su (蘇正煌)
- Department of Internal Medicine (Y.-J.W., C.-H.S., H.-I.Y.), MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan (Y.-J.W., C.-H.S., H.-H.W.)
| | - Hsueh-Hsiao Wang (王學孝)
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan (Y.-J.W., C.-H.S., H.-H.W.)
| | - Chin-Ling Hsieh (謝金玲)
- Department of Medical Research (T.-Y.T., C.-L.H., B.-J.W.), MacKay Memorial Hospital, Taipei, Taiwan
| | - Bo-Jeng Wang (王博正)
- Department of Medical Research (T.-Y.T., C.-L.H., B.-J.W.), MacKay Memorial Hospital, Taipei, Taiwan
| | - Yeu Su (蘇瑀)
- Institute of Biopharmaceutical Science/National Yang-Ming University, Taipei, Taiwan (T.-Y.T., Y.S.)
| | - Hung-I. Yeh (葉宏一)
- Department of Internal Medicine (Y.-J.W., C.-H.S., H.-I.Y.), MacKay Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
20
|
Schmidt K, de Wit C. Endothelium-Derived Hyperpolarizing Factor and Myoendothelial Coupling: The in vivo Perspective. Front Physiol 2021; 11:602930. [PMID: 33424626 PMCID: PMC7786115 DOI: 10.3389/fphys.2020.602930] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
The endothelium controls vascular tone adopting blood flow to tissue needs. It releases chemical mediators [e.g., nitric oxide (NO), prostaglandins (PG)] and exerts appreciable dilation through smooth muscle hyperpolarization, thus termed endothelium-dependent hyperpolarization (EDH). Initially, EDH was attributed to release of a factor, but later it was suggested that smooth muscle hyperpolarization might be derived from radial spread of an initial endothelial hyperpolarization through heterocellular channels coupling these vascular cells. The channels are indeed present and formed by connexins that enrich in gap junctions (GJ). In vitro data suggest that myoendothelial coupling underlies EDH-type dilations as evidenced by blocking experiments as well as simultaneous, merely identical membrane potential changes in endothelial and smooth muscle cells (SMCs), which is indicative of coupling through ohmic resistors. However, connexin-deficient animals do not display any attenuation of EDH-type dilations in vivo, and endothelial and SMCs exhibit distinct and barely superimposable membrane potential changes exerted by different means in vivo. Even if studied in the exact same artery EDH-type dilation exhibits distinct features in vitro and in vivo: in isometrically mounted vessels, it is rather weak and depends on myoendothelial coupling through connexin40 (Cx40), whereas in vivo as well as in vitro under isobaric conditions it is powerful and independent of myoendothelial coupling through Cx40. It is concluded that EDH-type dilations are distinct and a significant dependence on myoendothelial coupling in vitro does not reflect the situation under physiologic conditions in vivo. Myoendothelial coupling may act as a backup mechanism that is uncovered in the absence of the powerful EDH-type response and possibly reflects a situation in a pathophysiologic environment.
Collapse
Affiliation(s)
- Kjestine Schmidt
- Institut für Physiologie, Universität zu Lübeck, Lübeck, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Cor de Wit
- Institut für Physiologie, Universität zu Lübeck, Lübeck, Germany.,Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| |
Collapse
|
21
|
Abstract
Of the 21 members of the connexin family, 4 (Cx37, Cx40, Cx43, and Cx45) are expressed in the endothelium and/or smooth muscle of intact blood vessels to a variable and dynamically regulated degree. Full-length connexins oligomerize and form channel structures connecting the cytosol of adjacent cells (gap junctions) or the cytosol with the extracellular space (hemichannels). The different connexins vary mainly with regard to length and sequence of their cytosolic COOH-terminal tails. These COOH-terminal parts, which in the case of Cx43 are also translated as independent short isoforms, are involved in various cellular signaling cascades and regulate cell functions. This review focuses on channel-dependent and -independent effects of connexins in vascular cells. Channels play an essential role in coordinating and synchronizing endothelial and smooth muscle activity and in their interplay, in the control of vasomotor actions of blood vessels including endothelial cell reactivity to agonist stimulation, nitric oxide-dependent dilation, and endothelial-derived hyperpolarizing factor-type responses. Further channel-dependent and -independent roles of connexins in blood vessel function range from basic processes of vascular remodeling and angiogenesis to vascular permeability and interactions with leukocytes with the vessel wall. Together, these connexin functions constitute an often underestimated basis for the enormous plasticity of vascular morphology and function enabling the required dynamic adaptation of the vascular system to varying tissue demands.
Collapse
Affiliation(s)
- Ulrich Pohl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Planegg-Martinsried, Germany; Biomedical Centre, Cardiovascular Physiology, LMU Munich, Planegg-Martinsried, Germany; German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany; and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
22
|
Calhoun PJ, Phan AV, Taylor JD, James CC, Padget RL, Zeitz MJ, Smyth JW. Adenovirus targets transcriptional and posttranslational mechanisms to limit gap junction function. FASEB J 2020; 34:9694-9712. [PMID: 32485054 DOI: 10.1096/fj.202000667r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/05/2020] [Accepted: 05/12/2020] [Indexed: 01/19/2023]
Abstract
Adenoviruses are responsible for a spectrum of pathogenesis including viral myocarditis. The gap junction protein connexin43 (Cx43, gene name GJA1) facilitates rapid propagation of action potentials necessary for each heartbeat. Gap junctions also propagate innate and adaptive antiviral immune responses, but how viruses may target these structures is not understood. Given this immunological role of Cx43, we hypothesized that gap junctions would be targeted during adenovirus type 5 (Ad5) infection. We find reduced Cx43 protein levels due to decreased GJA1 mRNA transcripts dependent upon β-catenin transcriptional activity during Ad5 infection, with early viral protein E4orf1 sufficient to induce β-catenin phosphorylation. Loss of gap junction function occurs prior to reduced Cx43 protein levels with Ad5 infection rapidly inducing Cx43 phosphorylation events consistent with altered gap junction conductance. Direct Cx43 interaction with ZO-1 plays a critical role in gap junction regulation. We find loss of Cx43/ZO-1 complexing during Ad5 infection by co-immunoprecipitation and complementary studies in human induced pluripotent stem cell derived-cardiomyocytes reveal Cx43 gap junction remodeling by reduced ZO-1 complexing. These findings reveal specific targeting of gap junction function by Ad5 leading to loss of intercellular communication which would contribute to dangerous pathological states including arrhythmias in infected hearts.
Collapse
Affiliation(s)
- Patrick J Calhoun
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA.,Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Allen V Phan
- Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| | | | - Carissa C James
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA.,Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, USA
| | - Rachel L Padget
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA.,Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, USA
| | - Michael J Zeitz
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA
| | - James W Smyth
- Fralin Biomedical Research Institute at VTC, Roanoke, VA, USA.,Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA.,Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| |
Collapse
|
23
|
Hamard L, Santoro T, Allagnat F, Meda P, Nardelli-Haefliger D, Alonso F, Haefliger JA. Targeting connexin37 alters angiogenesis and arteriovenous differentiation in the developing mouse retina. FASEB J 2020; 34:8234-8249. [PMID: 32323401 DOI: 10.1096/fj.202000257r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/20/2020] [Accepted: 04/06/2020] [Indexed: 11/11/2022]
Abstract
Connexin37 (Cx37) forms intercellular channels between endothelial cells (EC), and contributes to coordinate the motor tone of vessels. We investigated the contribution of this protein during physiological angiogenesis. We show that, compared to WT littermates, mice lacking Cx37 (Cx37-/- ) featured (i) a decreased extension of the superficial vascular plexus during the first 4 days after birth; (ii) an increased vascular density at the angiogenic front at P6, due to an increase in the proliferative rate of EC and in the sprouting of the venous compartment, as well as to a somewhat displaced position of tip cells; (iii) a decreased coverage of newly formed arteries and veins by mural cells; (iv) altered ERK-dependent endothelial cells proliferation through the EphB4 signaling pathway, which is involved in the specification of veins and arteries. In vitro studies documented that, in the absence of Cx37, human venous EC (HUVEC) released less platelet-derived growth factor (PDGF) and more Angiopoietin-2, two molecules involved in the recruitment of mural cells. Treatment of mice with DAPT, an inhibitor of the Notch pathway, decreased the expression of Cx37, and partially mimicked in WT retinas, the alterations observed in Cx37-/- mice. Thus, Cx37 contributes to (i) the early angiogenesis of retina, by interacting with the Notch pathway; (ii) the growth and maturation of neo-vessels, by modulating tip, stalk, and mural cells; (iii) the regulation of arteriovenous specification, thus, representing a novel target for treatments of retina diseases.
Collapse
Affiliation(s)
- Lauriane Hamard
- Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Tania Santoro
- Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Florent Allagnat
- Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, Medical Center, University of Geneva, Geneva, Switzerland
| | | | - Florian Alonso
- Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), Université de Bordeaux, Bordeaux, France
| | | |
Collapse
|
24
|
Price GW, Chadjichristos CE, Kavvadas P, Tang SCW, Yiu WH, Green CR, Potter JA, Siamantouras E, Squires PE, Hills CE. Blocking Connexin-43 mediated hemichannel activity protects against early tubular injury in experimental chronic kidney disease. Cell Commun Signal 2020; 18:79. [PMID: 32450899 PMCID: PMC7249671 DOI: 10.1186/s12964-020-00558-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 03/23/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Tubulointerstitial fibrosis represents the key underlying pathology of Chronic Kidney Disease (CKD), yet treatment options remain limited. In this study, we investigated the role of connexin43 (Cx43) hemichannel-mediated adenosine triphosphate (ATP) release in purinergic-mediated disassembly of adherens and tight junction complexes in early tubular injury. METHODS Human primary proximal tubule epithelial cells (hPTECs) and clonal tubular epithelial cells (HK2) were treated with Transforming Growth Factor Beta1 (TGF-β1) ± apyrase, or ATPγS for 48 h. For inhibitor studies, cells were co-incubated with Cx43 mimetic Peptide 5, or purinergic receptor antagonists Suramin, A438079 or A804598. Immunoblotting, single-cell force spectroscopy and trans-epithelial electrical resistance assessed protein expression, cell-cell adhesion and paracellular permeability. Carboxyfluorescein uptake and biosensing measured hemichannel activity and real-time ATP release, whilst a heterozygous Cx43+/- mouse model with unilateral ureteral obstruction (UUO) assessed the role of Cx43 in vivo. RESULTS Immunohistochemistry of biopsy material from patients with diabetic nephropathy confirmed increased expression of purinergic receptor P2X7. TGF-β1 increased Cx43 mediated hemichannel activity and ATP release in hPTECs and HK2 cells. The cytokine reduced maximum unbinding forces and reduced cell-cell adhesion, which translated to increased paracellular permeability. Changes were reversed when cells were co-incubated with either Peptide 5 or P2-purinoceptor inhibitors. Cx43+/- mice did not exhibit protein changes associated with early tubular injury in a UUO model of fibrosis. CONCLUSION Data suggest that Cx43 mediated ATP release represents an initial trigger in early tubular injury via its actions on the adherens and tight junction complex. Since Cx43 is highly expressed in nephropathy, it represents a novel target for intervention of tubulointerstitial fibrosis in CKD. Video Abstract In proximal tubular epithelial cells (PTECs), tight junction proteins, including zona occuludens-1 (ZO-1), contribute to epithelial integrity, whilst the adherens junction protein epithelial (E)-cadherin (ECAD) maintains cell-cell coupling, facilitating connexin 43 (Cx43) gap junction-mediated intercellular communication (GJIC) and the direct transfer of small molecules and ions between cells. In disease, such as diabetic nephropathy, the pro-fibrotic cytokine transforming growth factor beta1 (TGF-β1) binds to its receptor and recruits SMAD2/3 signalling ahead of changes in gene transcription and up-regulation of Cx43-mediated hemichannels (HC). Uncoupled hemichannels permit the release of adenosine triphosphate (ATP) in to the extracellular space (↑[ATP]e), where ATP binds to the P2X7 purinoreceptor and activates the nucleotide-binding domain and leucine-rich repeat containing (NLR) protein-3 (NLRP3) inflammasome. Inflammation results in epithelial-to-mesenchymal transition (EMT), fibrosis and tubular injury. A major consequence is further loss of ECAD and reduced stickiness between cells, which can be functionally measured as a decrease in the maximum unbinding force needed to uncouple two adherent cells (Fmax). Loss of ECAD feeds forward to further lessen cell-cell coupling exacerbating the switch from GJIC to HC-mediated release of ATP. Reduction in ZO-1 impedes tight junction effectiveness and decreases trans-epithelial resistance (↓TER), resulting in increased paracellular permeability.
Collapse
Affiliation(s)
- Gareth W. Price
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Green Lane, Lincoln, UK
| | - Christos E. Chadjichristos
- National Institutes for Health and Medical Research Unite Mixte de Recherche S1155, Batiment Recherche, Tenon Hospital, 4 rue de la Chine, 75020 Paris, France
| | - Panagiotis Kavvadas
- National Institutes for Health and Medical Research Unite Mixte de Recherche S1155, Batiment Recherche, Tenon Hospital, 4 rue de la Chine, 75020 Paris, France
| | - Sydney C. W. Tang
- Division of Nephrology, Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Wai Han Yiu
- Division of Nephrology, Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Colin R. Green
- Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| | - Joe A. Potter
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Green Lane, Lincoln, UK
| | - Eleftherios Siamantouras
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Green Lane, Lincoln, UK
| | - Paul E. Squires
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Green Lane, Lincoln, UK
| | - Claire E. Hills
- Joseph Banks Laboratories, School of Life Sciences, University of Lincoln, Green Lane, Lincoln, UK
| |
Collapse
|
25
|
Angiotensin II induces RAW264.7 macrophage polarization to the M1‑type through the connexin 43/NF‑κB pathway. Mol Med Rep 2020; 21:2103-2112. [PMID: 32186758 PMCID: PMC7115186 DOI: 10.3892/mmr.2020.11023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 01/28/2020] [Indexed: 02/07/2023] Open
Abstract
Angiotensin II (AngII) serves an important inflammatory role in cardiovascular disease; it can induce macrophages to differentiate into the M1-type, produce inflammatory cytokines and resist pathogen invasion, and can cause a certain degree of damage to the body. Previous studies have reported that connexin 43 (Cx43) and NF-κB (p65) are involved in the AngII-induced inflammatory pathways of macrophages; however, the mechanisms underlying the effects of Cx43 and NF-κB (p65) on AngII-induced macrophage polarization have not been determined. Thus, the present study aimed to investigate the effects of Cx43 and NF-κB (p65) on the polarization process of AngII-induced macrophages. The macrophage polarization-related proteins and mRNAs were examined by flow cytometry, western blotting, immunofluorescence, ELISA and reverse transcription-quantitative PCR analyses. RAW264.7 macrophages were treated with AngII to simulate chronic inflammation and it was subsequently found that AngII promoted RAW 264.7 macrophage polarization towards the M1-type by such effects as the release of inducible nitric oxide synthase (iNOS), tumour necrosis factor (TNF)-α, IL-1β, the secretion of IL-6, and the expression of M1-type indicators, such as CD86. Simultaneously, compared with the control group, the protein expression levels of Cx43 and phosphorylated (p)-p65 were significantly increased following AngII treatment. The M1-related phenotypic indicators, iNOS, TNF-α, IL-1β, IL-6 and CD86, were inhibited by the NF-κB (p65) signalling pathway inhibitor BAY117082. Similarly, the Cx43 inhibitors, Gap26 and Gap19, also inhibited the expression of M1-related factors, and the protein expression levels of p-p65 in the Gap26/Gap19 groups were significantly decreased compared with the AngII group. Altogether, these findings suggested that AngII may induce the polarization of RAW264.7 macrophages to the M1-type through the Cx43/NF-κB (p65) signalling pathway.
Collapse
|
26
|
Lagos-Cabré R, Burgos-Bravo F, Avalos AM, Leyton L. Connexins in Astrocyte Migration. Front Pharmacol 2020; 10:1546. [PMID: 32009957 PMCID: PMC6974553 DOI: 10.3389/fphar.2019.01546] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/29/2019] [Indexed: 12/18/2022] Open
Abstract
Astrocytes have long been considered the supportive cells of the central nervous system, but during the last decades, they have gained much more attention because of their active participation in the modulation of neuronal function. For example, after brain damage, astrocytes become reactive and undergo characteristic morphological and molecular changes, such as hypertrophy and increase in the expression of glial fibrillary acidic protein (GFAP), in a process known as astrogliosis. After severe damage, astrocytes migrate to the lesion site and proliferate, which leads to the formation of a glial scar. At this scar-forming stage, astrocytes secrete many factors, such as extracellular matrix proteins, cytokines, growth factors and chondroitin sulfate proteoglycans, stop migrating, and the process is irreversible. Although reactive gliosis is a normal physiological response that can protect brain cells from further damage, it also has detrimental effects on neuronal survival, by creating a hostile and non-permissive environment for axonal repair. The transformation of astrocytes from reactive to scar-forming astrocytes highlights migration as a relevant regulator of glial scar formation, and further emphasizes the importance of efficient communication between astrocytes in order to orchestrate cell migration. The coordination between astrocytes occurs mainly through Connexin (Cx) channels, in the form of direct cell-cell contact (gap junctions, GJs) or contact between the extracellular matrix and the astrocytes (hemichannels, HCs). Reactive astrocytes increase the expression levels of several proteins involved in astrocyte migration, such as αvβ3 Integrin, Syndecan-4 proteoglycan, the purinergic receptor P2X7, Pannexin1, and Cx43 HCs. Evidence has indicated that Cx43 HCs play a role in regulating astrocyte migration through the release of small molecules to the extracellular space, which then activate receptors in the same or adjacent cells to continue the signaling cascades required for astrocyte migration. In this review, we describe the communication of astrocytes through Cxs, the role of Cxs in inflammation and astrocyte migration, and discuss the molecular mechanisms that regulate Cx43 HCs, which may provide a therapeutic window of opportunity to control astrogliosis and the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Raúl Lagos-Cabré
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Center for Studies on Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Francesca Burgos-Bravo
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Center for Studies on Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Ana María Avalos
- Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Center for Studies on Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| |
Collapse
|
27
|
McArthur L, Riddell A, Chilton L, Smith GL, Nicklin SA. Regulation of connexin 43 by interleukin 1β in adult rat cardiac fibroblasts and effects in an adult rat cardiac myocyte: fibroblast co-culture model. Heliyon 2019; 6:e03031. [PMID: 31909243 PMCID: PMC6940628 DOI: 10.1016/j.heliyon.2019.e03031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/22/2019] [Accepted: 12/10/2019] [Indexed: 01/05/2023] Open
Abstract
Connexin 43 expression (Cx43) is increased in cardiac fibroblasts (CFs) following myocardial infarction. Here, potential mediators responsible for increasing Cx43 expression and effects of differential CF phenotype on cardiac myocyte (CM) function were investigated. Stimulating adult rat CFs with proinflammatory mediators revealed that interleukin 1β (IL-1β) significantly enhanced Cx43 levels through the IL-1β pathway. Additionally, IL-1β reduced mRNA levels of the myofibroblast (MF) markers: (i) connective tissue growth factor (CTGF) and (ii) α smooth muscle actin (αSMA), compared to control CFs. A co-culture adult rat CM:CF model was utilised to examine cell-to-cell interactions. Transfer of calcein from CMs to underlying CFs suggested functional gap junction formation. Functional analysis revealed contraction duration (CD) of CMs was shortened in co-culture with CFs, while treatment of CFs with IL-1β reduced this mechanical effect of co-culture. No effect on action potential rise time or duration of CMs cultured with control or IL-1β-treated CFs was observed. These data demonstrate that stimulating CFs with IL-1β increases Cx43 and reduces MF marker expression, suggesting altered cell phenotype. These changes may underlie the reduced mechanical effects of IL-1β treated CFs on CD of co-cultured CMs and therefore have an implication for our understanding of heterocellular interactions in cardiac disease.
Collapse
Affiliation(s)
- Lisa McArthur
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Alexandra Riddell
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Lisa Chilton
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Queensland, Australia
| | - Godfrey L Smith
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Stuart A Nicklin
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| |
Collapse
|
28
|
Latorre M, Bersi MR, Humphrey JD. Computational Modeling Predicts Immuno-Mechanical Mechanisms of Maladaptive Aortic Remodeling in Hypertension. INTERNATIONAL JOURNAL OF ENGINEERING SCIENCE 2019; 141:35-46. [PMID: 32831391 PMCID: PMC7437922 DOI: 10.1016/j.ijengsci.2019.05.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Uncontrolled hypertension is a major risk factor for myriad cardiovascular diseases. Among its many effects, hypertension increases central artery stiffness which in turn is both an initiator and indicator of disease. Despite extensive clinical, animal, and basic science studies, the biochemomechanical mechanisms by which hypertension drives aortic stiffening remain unclear. In this paper, we show that a new computational model of aortic growth and remodeling can capture differential effects of induced hypertension on the thoracic and abdominal aorta in a common mouse model of disease. Because the simulations treat the aortic wall as a constrained mixture of different constituents having different material properties and rates of turnover, one can gain increased insight into underlying constituent-level mechanisms of aortic remodeling. Model results suggest that the aorta can mechano-adapt locally to blood pressure elevation in the absence of marked inflammation, but large increases in inflammation drive a persistent maladaptive phenotype characterized primarily by adventitial fibrosis. Moreover, this fibrosis appears to occur via a marked increase in the rate of deposition of collagen having different material properties in the absence of a compensatory increase in the rate of matrix degradation. Controlling inflammation thus appears to be key to reducing fibrosis, but therapeutic strategies should not compromise the proteolytic activity of the wall that is essential to mechanical homeostasis.
Collapse
Affiliation(s)
- Marcos Latorre
- Department of Biomedical Engineering Yale University, New Haven, CT, USA
| | - Matthew R. Bersi
- Department of Biomedical Engineering Vanderbilt University, Nashville, TN, USA
| | - Jay D. Humphrey
- Department of Biomedical Engineering Yale University, New Haven, CT, USA
- Vascular Biology and Therapeutics Program Yale School of Medicine, New Haven, CT, USA
- Corresponding author: (Jay D. Humphrey)
| |
Collapse
|
29
|
Wang M, Wu Y, Yu Y, Fu Y, Yan H, Wang X, Li T, Peng W, Luo D. Rutaecarpine prevented ox-LDL-induced VSMCs dysfunction through inhibiting overexpression of connexin 43. Eur J Pharmacol 2019; 853:84-92. [DOI: 10.1016/j.ejphar.2019.03.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/14/2019] [Accepted: 03/14/2019] [Indexed: 01/29/2023]
|
30
|
Le Gal L, Pellegrin M, Santoro T, Mazzolai L, Kurtz A, Meda P, Wagner C, Haefliger J. Connexin37-Dependent Mechanisms Selectively Contribute to Modulate Angiotensin II -Mediated Hypertension. J Am Heart Assoc 2019; 8:e010823. [PMID: 30943815 PMCID: PMC6507190 DOI: 10.1161/jaha.118.010823] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/30/2019] [Indexed: 12/23/2022]
Abstract
Background Gap junction channels made of Connexin37 (Cx37) are expressed by aortic endothelial and smooth muscle cells of hypertensive mice, as well as by the renin-secreting cells of kidneys. Methods and Results To decipher whether Cx37 has any role in hypertension, angiotensin II (Ang II ) was infused in normotensive wild-type and Cx37-deficient mice (Cx37-/-). After 2 to 4 weeks, the resulting increase in blood pressure was lower in Cx37-/- than in wild-type mice, suggesting an alteration in the Ang II response. To investigate this possibility, mice were submitted to a 2-kidney, 1-clip procedure, a renin-dependent model of hypertension. Two weeks after this clipping, Cx37-/- mice were less hypertensive than wild-type mice and, 2 weeks later, their blood pressure had returned to control values, in spite of abnormally high plasma renin levels. In contrast, Cx37-/- and wild-type mice that received N-nitro-l-arginine-methyl-ester, a renin-independent model of hypertension, featured a similar and sustained increase in blood pressure. The data indicate that loss of Cx37 selectively altered the Ang II -dependent pathways. Consistent with this conclusion, aortas of Cx37-/- mice featured an increased basal expression of the Ang II type 2 receptors ( AT 2R), and increased transcripts levels of downstream signaling proteins, such as Cnksr1 and Ptpn6 ( SHP -1). Accordingly, the response of Cx37-/- mice aortas to an ex vivo Ang II exposure was altered, since phosphorylation levels of several proteins of the Ang II pathway ( MLC 2, ERK , and AKT ) remained unchanged. Conclusions These findings provide evidence that Cx37 selectively influences Ang II signaling, mostly via a modulation of the expression of the Ang II type 2 receptor.
Collapse
Affiliation(s)
- Loïc Le Gal
- Department of MedicineUniversity of LausanneSwitzerland
| | - Maxime Pellegrin
- Division of AngiologyHeart and Vessel DepartmentCentre Hospitalier Universitaire VaudoisUniversity of LausanneSwitzerland
| | - Tania Santoro
- Department of MedicineUniversity of LausanneSwitzerland
| | - Lucia Mazzolai
- Division of AngiologyHeart and Vessel DepartmentCentre Hospitalier Universitaire VaudoisUniversity of LausanneSwitzerland
| | - Armin Kurtz
- Department of PhysiologyUniversity of RegensburgGermany
| | - Paolo Meda
- Department of Cell Physiology and MetabolismSchool of MedicineCMUUniversity of GenevaSwitzerland
| | | | | |
Collapse
|
31
|
Longchamp A, Mirabella T, Arduini A, MacArthur MR, Das A, Treviño-Villarreal JH, Hine C, Ben-Sahra I, Knudsen NH, Brace LE, Reynolds J, Mejia P, Tao M, Sharma G, Wang R, Corpataux JM, Haefliger JA, Ahn KH, Lee CH, Manning BD, Sinclair DA, Chen CS, Ozaki CK, Mitchell JR. Amino Acid Restriction Triggers Angiogenesis via GCN2/ATF4 Regulation of VEGF and H 2S Production. Cell 2019; 173:117-129.e14. [PMID: 29570992 DOI: 10.1016/j.cell.2018.03.001] [Citation(s) in RCA: 224] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/17/2018] [Accepted: 02/27/2018] [Indexed: 12/15/2022]
Abstract
Angiogenesis, the formation of new blood vessels by endothelial cells (ECs), is an adaptive response to oxygen/nutrient deprivation orchestrated by vascular endothelial growth factor (VEGF) upon ischemia or exercise. Hypoxia is the best-understood trigger of VEGF expression via the transcription factor HIF1α. Nutrient deprivation is inseparable from hypoxia during ischemia, yet its role in angiogenesis is poorly characterized. Here, we identified sulfur amino acid restriction as a proangiogenic trigger, promoting increased VEGF expression, migration and sprouting in ECs in vitro, and increased capillary density in mouse skeletal muscle in vivo via the GCN2/ATF4 amino acid starvation response pathway independent of hypoxia or HIF1α. We also identified a requirement for cystathionine-γ-lyase in VEGF-dependent angiogenesis via increased hydrogen sulfide (H2S) production. H2S mediated its proangiogenic effects in part by inhibiting mitochondrial electron transport and oxidative phosphorylation, resulting in increased glucose uptake and glycolytic ATP production.
Collapse
Affiliation(s)
- Alban Longchamp
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Surgery and the Heart and Vascular Center, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Teodelinda Mirabella
- Tissue Microfabrication Lab, Department of Biomedical Engineering, Boston University, Boston, MA, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA, USA
| | - Alessandro Arduini
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Michael R MacArthur
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Abhirup Das
- Glenn Center for the Biological Mechanisms of Aging, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Laboratory for Ageing Research, Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney NSW 2052, Australia
| | | | - Christopher Hine
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Issam Ben-Sahra
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Nelson H Knudsen
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Lear E Brace
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Justin Reynolds
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Pedro Mejia
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ming Tao
- Department of Surgery and the Heart and Vascular Center, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Gaurav Sharma
- Department of Surgery and the Heart and Vascular Center, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Rui Wang
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, ON, Canada
| | - Jean-Marc Corpataux
- Department of Vascular Surgery, Laboratory of Experimental Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Jacques-Antoine Haefliger
- Department of Vascular Surgery, Laboratory of Experimental Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Kyo Han Ahn
- Department of Chemistry, Postech, 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, Republic of Korea
| | - Chih-Hao Lee
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Brendan D Manning
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - David A Sinclair
- Glenn Center for the Biological Mechanisms of Aging, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Laboratory for Ageing Research, Department of Pharmacology, School of Medical Sciences, University of New South Wales, Sydney NSW 2052, Australia
| | - Christopher S Chen
- Tissue Microfabrication Lab, Department of Biomedical Engineering, Boston University, Boston, MA, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA, USA
| | - C Keith Ozaki
- Department of Surgery and the Heart and Vascular Center, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - James R Mitchell
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
32
|
Cao L, Zhao C, Cong H, Hou K, Wan L, Wang J, Zhao L, Yan H. The effect of Telmisartan on the expression of connexin43 and neointimal hyperplasia in a rabbit iliac artery restenosis model. Heart Vessels 2019; 34:1230-1239. [PMID: 30671641 DOI: 10.1007/s00380-018-01338-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/28/2018] [Indexed: 10/27/2022]
Abstract
We established a rabbit iliac artery restenosis model to explore the impact of Telmisartan on the expression of Connexin43 (Cx43) and neointimal hyperplasia. Thirty New Zealand white rabbits were randomly divided into three groups: control group (n = 10), restenosis group (n = 10), and Telmisartan group (n = 10). The restenosis model was established by high-cholesterol diet combined with double-balloon injury of iliac arteries. In addition, Telmisartan at 5 mg/(kg day) was administered to the rabbits of Telmisartan group on the second day after the second balloon injury. All rabbits were killed at the end of the experiment followed by institution policy. Before sacrifice, blood samples were obtained to test serum angiotensinII (AngII). Iliac arteries were isolated for morphological analysis and determining the expression of Cx43 by HE staining, immunohistochemical analysis, reverse transcription-polymerase chain reaction (RT-PCR), and Western Blotting analysis. Then, the local AngII levels of arteries were measured by radioimmunoassay. As compared with controls, the expression of Cx43 mRNA (0.98 ± 0.08) vs. (1.27 ± 0.17), P < 0.01), and Cx43 protein [(0.75 ± 0.08) vs. (0.90 ± 0.08), P < 0.05] of restenosis group were increased, which were significantly higher than those of Telmisartan group [Cx43 mRNA: (1.27 ± 0.17) vs. (1.00 ± 0.20), P < 0.01; Cx43 protein: (0.90 ± 0.08) vs. (0.82 ± 0.05), P < 0.05]. Furthermore, The intima thickness [(266.12 ± 70.27) vs. (2.85 ± 0.19) μm, P < 0.01] and the local AngII [(115.6 ± 15.7) vs. (90.1 ± 7.7), P < 0.05] of restenosis group were raised when compared with controls. Telmisartan group exhibited thinner intima compared with restenosis group [(68.22 ± 24.37) vs. (266.12 ± 70.27), P < 0.01]. However, the local AngII levels between these two groups were approximate. In addition, the plasma concentration of AngII was not significantly different among three groups. In conclusion, Telmisartan can inhibit the expression of connexin43 and neointimal hyperplasia in iliac artery restenosis model.
Collapse
Affiliation(s)
- Lu Cao
- Cardiology Department, Tianjin Chest Hospital, Tianjin, 300222, People's Republic of China.
| | - Cui Zhao
- National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, People's Republic of China
| | - Hongliang Cong
- Cardiology Department, Tianjin Chest Hospital, Tianjin, 300222, People's Republic of China
| | - Kai Hou
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
| | - Lianghui Wan
- Cardiology Department, Tianjin Chest Hospital, Tianjin, 300222, People's Republic of China
| | - Jixiang Wang
- Cardiology Department, Tianjin Chest Hospital, Tianjin, 300222, People's Republic of China
| | - Lili Zhao
- Cardiology Department, Tianjin Chest Hospital, Tianjin, 300222, People's Republic of China
| | - Haiyang Yan
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Affiliated Hospital of Logistics University of the Chinese People's Armed Police Forces, Tianjin, 300162, People's Republic of China
| |
Collapse
|
33
|
ERK and miRNA-1 target Cx43 expression and phosphorylation to modulate the vascular protective effect of angiotensin II. Life Sci 2019; 216:59-66. [DOI: 10.1016/j.lfs.2018.11.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/01/2018] [Accepted: 11/08/2018] [Indexed: 12/21/2022]
|
34
|
Zhao X, Wang X. Candesartan targeting of angiotensin II type 1 receptor demonstrates benefits for hypertension in pregnancy via the NF‑κB signaling pathway. Mol Med Rep 2018; 18:705-714. [PMID: 29845261 PMCID: PMC6059699 DOI: 10.3892/mmr.2018.9070] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 12/13/2017] [Indexed: 12/19/2022] Open
Abstract
Hypertensive disorders may be a complication of pregnancy and are characterized by the high blood pressure. Evidence suggests that alterations in the renin-angiotensin-aldosterone system and the sympathetic nervous system are associated with gestational hypertension. Angiotensin II type 1 receptor (Ang-IITR) is a potential target in the progression of gestational hypertension. Candesartan is selective Ang-IITR antagonist that may act against vasoconstriction and reduces peripheral vascular resistance. The aim of the present study was to evaluate the efficacy of Candesartan and the underlying molecular mechanism of the nuclear factor-κB (NF-κB) signaling pathway in the progression of gestational hypertension in a mouse model. Expression and activity of Ang-IITR was evaluated in a mouse model of gestational hypertension prior to and post-treatment of Candesartan both in vitro and in vivo. It was determined whether Candesartan treatment reduces higher blood pressure activated the renal renin-angiotensin system and a prognostic marker, soluble endoglin, and its associated gene in mice with gestational hypertension. Angiotensin-converting enzyme plasma levels and activity were also evaluated in the present study. Cytoplasmic and nuclear immunostaining of NF-κB and associated proteins transforming growth factor β (TGF-β) and endoglin was enhanced in vascular endothelial cells and mice with gestational hypertension. Soluble fms-like tyrosine kinase 1 (sFlt-1), insulin resistance homeostasis model assessment score and associated cardiovascular risk factors also were measured. Results demonstrated that angiotensin and Ang-IITR expression levels were upregulated in mice with gestational hypertension and were downregulated by Candesartan treatment. Renal renin-angiotensin and soluble endoglin were also improved in mice in the Candesartan-treated group. In addition, Candesartan treatment enhanced NF-κB activity, as well as TGF-β and vascular endothelial growth factor expression which led to improved levels of sFlt-1, insulin resistance homeostasis and associated cardiovascular risk factors. Gestational hypertension was markedly improved by treatment of Candesartan compared with the control. In conclusion, the findings of the present study suggested that the NF-κB signaling pathway may be involved in with Candesartan-mediated Ang-IITR for the treatment of gestational hypertension.
Collapse
Affiliation(s)
- Xudong Zhao
- Department of Gynecology and Obstetrics, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Xietong Wang
- Department of Gynecology and Obstetrics, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
35
|
Mishra JS, More AS, Hankins GDV, Kumar S. Hyperandrogenemia reduces endothelium-derived hyperpolarizing factor-mediated relaxation in mesenteric artery of female rats. Biol Reprod 2018; 96:1221-1230. [PMID: 28486649 DOI: 10.1093/biolre/iox043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/08/2017] [Indexed: 01/05/2023] Open
Abstract
Women with polycystic ovary syndrome (PCOS) are often presented with hyperandrogenemia along with vascular dysfunction and elevated blood pressure. In animal models of PCOS, anti-androgen treatment decreased blood pressure, indicating a key role for androgens in the development of hypertension. However, the underlying androgen-mediated mechanism that contributes to increased blood pressure is not known. This study determined whether elevated androgens affect endothelium-derived hyperpolarizing factor (EDHF)-mediated vascular relaxation responses through alteration in function of gap junctional proteins. Female rats were implanted with placebo or dihydrotestosterone (DHT) pellets (7.5 mg, 90-day release). After 12 weeks of DHT exposure, blood pressure was assessed through carotid arterial catheter and endothelium-dependent mesenteric arterial EDHF relaxation using wire myograph. Connexin expression in mesenteric arteries was also examined. Elevated DHT significantly increased mean arterial pressure and decreased endothelium-dependent EDHF-mediated acetylcholine relaxation. Inhibition of Cx40 did not have any effect, while inhibition of Cx37 decreased EDHF relaxation to a similar magnitude in both controls and DHT females. On the other hand, inhibition of Cx43 significantly attenuated EDHF relaxation in mesenteric arteries of controls but not DHT females. Elevated DHT did not alter Cx37 or Cx40, but decreased Cx43 mRNA and protein levels in mesenteric arteries. In vitro exposure of DHT to cultured mesenteric artery smooth muscle cells dose-dependently downregulated Cx43 expression. In conclusion, increased blood pressure in hyperandrogenic females is due, at least in part, to decreased EDHF-mediated vascular relaxation responses. Decreased Cx43 expression and activity may play a role in contributing to androgen-induced decrease in EDHF function.
Collapse
Affiliation(s)
- Jay S Mishra
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Texas, USA
| | - Amar S More
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Texas, USA
| | - Gary D V Hankins
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Texas, USA
| | - Sathish Kumar
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Texas, USA
| |
Collapse
|
36
|
Li J, Wang S, Bai J, Yang XL, Zhang YL, Che YL, Li HH, Yang YZ. Novel Role for the Immunoproteasome Subunit PSMB10 in Angiotensin II-Induced Atrial Fibrillation in Mice. Hypertension 2018; 71:866-876. [PMID: 29507100 DOI: 10.1161/hypertensionaha.117.10390] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 10/14/2017] [Accepted: 02/11/2018] [Indexed: 11/16/2022]
Abstract
Angiotensin II (Ang II) and inflammation are associated with pathogenesis of atrial fibrillation (AF), but the underlying molecular mechanisms of these events remain unknown. The immunoproteasome has emerged as a critical regulator of inflammatory responses. Here, we investigated its role in Ang II-induced AF in immunosubunit PSMB10 (also known as β2i or LMP10) knockout (KO) mice. AF was induced by Ang II infusion (2000 ng/min per kg). PSMB10 expression and trypsin-like activity were increased in atrial tissues and serum from Ang II-treated mice or serum from patients with AF. Moreover, Ang II-infused wild-type (WT) mice had a higher AF and increased atrial fibrosis, reactive oxygen species production, and inflammation compared with saline-treated WT animals. These effects were attenuated in PSMB10 KO mice but were aggravated in recombinant adeno-associated virus serotype 9-PSMB10-treated mice. Administration of IKKβ-specific inhibitor IMD 0354 reduced Ang II-induced AF, reactive oxygen species production, inflammation, and NF-kB (nuclear factor-kB) activation. Mechanistically, Ang II infusion upregulated PSMB10 expression to promote PTEN (phosphatase and tensin homolog deleted on chromosome ten) degradation and AKT1 activation, which not only activated TGF-β-Smad2/3 signaling leading to cardiac fibrosis but also induced IKKβ activation and ubiquitin-mediated degradation of IkBα ultimately resulting in activation of NF-kB target genes (IL [interleukin]-1β, IL-6, NOX [NADPH oxidase] 2, NOX4, and CX43 [connexin 43]). Overall, our study identifies immunosubunit PSMB10 as a novel regulator that contributes to Ang II-induced AF and suggests that inhibition of PSMB10 may represent a potential therapeutic target for treating hypertensive AF.
Collapse
Affiliation(s)
- Jing Li
- From the Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, China (J.L., X.-L.Y., Y.-L.Z., Y.-Z.Y., H.-H.L.); Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, China (J.B., H.-H.L.); and Department of Ophthalmology (S.W.) and Department of Radiotherapy Oncology (Y.-L.C.), The Second Affiliated Hospital of Dalian Medical University, China
| | - Shuai Wang
- From the Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, China (J.L., X.-L.Y., Y.-L.Z., Y.-Z.Y., H.-H.L.); Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, China (J.B., H.-H.L.); and Department of Ophthalmology (S.W.) and Department of Radiotherapy Oncology (Y.-L.C.), The Second Affiliated Hospital of Dalian Medical University, China
| | - Jie Bai
- From the Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, China (J.L., X.-L.Y., Y.-L.Z., Y.-Z.Y., H.-H.L.); Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, China (J.B., H.-H.L.); and Department of Ophthalmology (S.W.) and Department of Radiotherapy Oncology (Y.-L.C.), The Second Affiliated Hospital of Dalian Medical University, China
| | - Xiao-Lei Yang
- From the Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, China (J.L., X.-L.Y., Y.-L.Z., Y.-Z.Y., H.-H.L.); Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, China (J.B., H.-H.L.); and Department of Ophthalmology (S.W.) and Department of Radiotherapy Oncology (Y.-L.C.), The Second Affiliated Hospital of Dalian Medical University, China
| | - Yun-Long Zhang
- From the Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, China (J.L., X.-L.Y., Y.-L.Z., Y.-Z.Y., H.-H.L.); Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, China (J.B., H.-H.L.); and Department of Ophthalmology (S.W.) and Department of Radiotherapy Oncology (Y.-L.C.), The Second Affiliated Hospital of Dalian Medical University, China
| | - Yi-Lin Che
- From the Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, China (J.L., X.-L.Y., Y.-L.Z., Y.-Z.Y., H.-H.L.); Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, China (J.B., H.-H.L.); and Department of Ophthalmology (S.W.) and Department of Radiotherapy Oncology (Y.-L.C.), The Second Affiliated Hospital of Dalian Medical University, China
| | - Hui-Hua Li
- From the Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, China (J.L., X.-L.Y., Y.-L.Z., Y.-Z.Y., H.-H.L.); Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, China (J.B., H.-H.L.); and Department of Ophthalmology (S.W.) and Department of Radiotherapy Oncology (Y.-L.C.), The Second Affiliated Hospital of Dalian Medical University, China.
| | - Yan-Zong Yang
- From the Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, China (J.L., X.-L.Y., Y.-L.Z., Y.-Z.Y., H.-H.L.); Department of Nutrition and Food Hygiene, School of Public Health, Dalian Medical University, China (J.B., H.-H.L.); and Department of Ophthalmology (S.W.) and Department of Radiotherapy Oncology (Y.-L.C.), The Second Affiliated Hospital of Dalian Medical University, China.
| |
Collapse
|
37
|
Looft-Wilson RC, Goodell CR, Mutch CA, Mutchler SM, Miller KL, Guraya M. Increased myoendothelial feedback is associated with increased connexin37 and IK1 channel expression in mesenteric arteries of diet-induced hyperhomocysteinemic mice. Microcirculation 2017; 24. [PMID: 28857417 DOI: 10.1111/micc.12398] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 08/24/2017] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Previously, we found that diet-induced HHcy in mice caused decreased eNOS expression and signaling in mesenteric arteries, but greatly enhanced non-NOS, non-prostacyclin-dependent vasodilation, which involves MEJ communication. To further assess whether HHcy enhances MEJ communication, this study examined endothelium-dependent attenuation of phenylephrine-induced vasoconstriction (myoendothelial feedback) and key molecules involved. METHODS Myoendothelial feedback was examined in isolated mouse mesenteric arteries, after 6-weeks diet-induced HHcy, using pressure myography. Gap junction (Cx37, Cx40, Cx43), NOS (eNOS, nNOS, iNOS), and potassium channel (IK1) protein expression were measured with immunoblots, and connexin mRNAs with real-time PCR. Contribution of nNOS + iNOS to vasomotor responses was assessed using the drug TRIM. RESULTS Myoendothelial feedback was significantly (P < .05) enhanced in HHcy arteries compared to control, coincident with significantly greater Cx37 and IK1 protein and Cx37 mRNA. Cx43 protein, but not mRNA, was significantly less in HHcy, and Cx40 was not different. eNOS protein was significantly less in HHcy. nNOS and iNOS were not different. TRIM had little effect on vasomotor function. CONCLUSIONS Diet-induced HHcy enhanced myoendothelial feedback, and increased Cx37 and IK1 expression may contribute. nNOS or iNOS did not upregulate to compensate for decreased eNOS, and they had little involvement in vasomotor function.
Collapse
Affiliation(s)
- Robin C Looft-Wilson
- Department of Kinesiology and Health Sciences, The College of William & Mary, Williamsburg, VA, USA
| | - Cara R Goodell
- Department of Kinesiology and Health Sciences, The College of William & Mary, Williamsburg, VA, USA
| | - Christina A Mutch
- Department of Kinesiology and Health Sciences, The College of William & Mary, Williamsburg, VA, USA
| | - Stephanie M Mutchler
- Department of Kinesiology and Health Sciences, The College of William & Mary, Williamsburg, VA, USA
| | - Kayla L Miller
- Department of Kinesiology and Health Sciences, The College of William & Mary, Williamsburg, VA, USA
| | - Monique Guraya
- Department of Kinesiology and Health Sciences, The College of William & Mary, Williamsburg, VA, USA
| |
Collapse
|
38
|
Haefliger JA, Allagnat F, Hamard L, Le Gal L, Meda P, Nardelli-Haefliger D, Génot E, Alonso F. Targeting Cx40 (Connexin40) Expression or Function Reduces Angiogenesis in the Developing Mouse Retina. Arterioscler Thromb Vasc Biol 2017; 37:2136-2146. [PMID: 28982669 DOI: 10.1161/atvbaha.117.310072] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/20/2017] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Cx40 (Connexin40) forms intercellular channels that coordinate the electric conduction in the heart and the vasomotor tone in large vessels. The protein was shown to regulate tumoral angiogenesis; however, whether Cx40 also contributes to physiological angiogenesis is still unknown. APPROACH AND RESULTS Here, we show that Cx40 contributes to physiological angiogenesis. Genetic deletion of Cx40 leads to a reduction in vascular growth and capillary density in the neovascularization model of the mouse neonatal retina. At the angiogenic front, vessel sprouting is reduced, and the mural cells recruited along the sprouts display an altered phenotype. These alterations can be attributed to disturbed endothelial cell functions as selective reexpression of Cx40 in these cells restores normal angiogenesis. In vitro, targeting Cx40 in microvascular endothelial cells, by silencing its expression or by blocking gap junction channels, decreases their proliferation. Moreover, loss of Cx40 in these cells also increases their release of PDGF (platelet-derived growth factor) and promotes the chemoattraction of mural cells. In vivo, an intravitreal injection of a Cx40 inhibitory peptide, phenocopies the loss of Cx40 in the retinal vasculature of wild-type mice. CONCLUSIONS Collectively, our data show that endothelial Cx40 contributes to the early stages of physiological angiogenesis in the developing retina, by regulating vessel growth and maturation. Cx40 thus represents a novel therapeutic target for treating pathological ocular angiogenesis.
Collapse
Affiliation(s)
- Jacques-Antoine Haefliger
- From the Department of Medicine (J.-A.H., F.A., L.H., L.L.G., F.A.) and Department of Urology (D.N.H.), Lausanne University Hospital, Switzerland; Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Switzerland (P.M.); and Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), Université de Bordeaux, France (E.G., F.A.).
| | - Florent Allagnat
- From the Department of Medicine (J.-A.H., F.A., L.H., L.L.G., F.A.) and Department of Urology (D.N.H.), Lausanne University Hospital, Switzerland; Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Switzerland (P.M.); and Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), Université de Bordeaux, France (E.G., F.A.)
| | - Lauriane Hamard
- From the Department of Medicine (J.-A.H., F.A., L.H., L.L.G., F.A.) and Department of Urology (D.N.H.), Lausanne University Hospital, Switzerland; Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Switzerland (P.M.); and Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), Université de Bordeaux, France (E.G., F.A.)
| | - Loïc Le Gal
- From the Department of Medicine (J.-A.H., F.A., L.H., L.L.G., F.A.) and Department of Urology (D.N.H.), Lausanne University Hospital, Switzerland; Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Switzerland (P.M.); and Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), Université de Bordeaux, France (E.G., F.A.)
| | - Paolo Meda
- From the Department of Medicine (J.-A.H., F.A., L.H., L.L.G., F.A.) and Department of Urology (D.N.H.), Lausanne University Hospital, Switzerland; Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Switzerland (P.M.); and Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), Université de Bordeaux, France (E.G., F.A.)
| | - Denise Nardelli-Haefliger
- From the Department of Medicine (J.-A.H., F.A., L.H., L.L.G., F.A.) and Department of Urology (D.N.H.), Lausanne University Hospital, Switzerland; Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Switzerland (P.M.); and Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), Université de Bordeaux, France (E.G., F.A.)
| | - Elisabeth Génot
- From the Department of Medicine (J.-A.H., F.A., L.H., L.L.G., F.A.) and Department of Urology (D.N.H.), Lausanne University Hospital, Switzerland; Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Switzerland (P.M.); and Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), Université de Bordeaux, France (E.G., F.A.)
| | - Florian Alonso
- From the Department of Medicine (J.-A.H., F.A., L.H., L.L.G., F.A.) and Department of Urology (D.N.H.), Lausanne University Hospital, Switzerland; Department of Cell Physiology and Metabolism, University of Geneva Medical Center, Switzerland (P.M.); and Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), Université de Bordeaux, France (E.G., F.A.).
| |
Collapse
|
39
|
Lagos-Cabré R, Alvarez A, Kong M, Burgos-Bravo F, Cárdenas A, Rojas-Mancilla E, Pérez-Nuñez R, Herrera-Molina R, Rojas F, Schneider P, Herrera-Marschitz M, Quest AFG, van Zundert B, Leyton L. α Vβ 3 Integrin regulates astrocyte reactivity. J Neuroinflammation 2017; 14:194. [PMID: 28962574 PMCID: PMC5622429 DOI: 10.1186/s12974-017-0968-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 09/20/2017] [Indexed: 12/14/2022] Open
Abstract
Background Neuroinflammation involves cytokine release, astrocyte reactivity and migration. Neuronal Thy-1 promotes DITNC1 astrocyte migration by engaging αVβ3 Integrin and Syndecan-4. Primary astrocytes express low levels of these receptors and are unresponsive to Thy-1; thus, inflammation and astrocyte reactivity might be necessary for Thy-1-induced responses. Methods Wild-type rat astrocytes (TNF-activated) or from human SOD1G93A transgenic mice (a neurodegenerative disease model) were used to evaluate cell migration, Thy-1 receptor levels, signaling molecules, and reactivity markers. Results Thy-1 induced astrocyte migration only after TNF priming. Increased expression of αVβ3 Integrin, Syndecan-4, P2X7R, Pannexin-1, Connexin-43, GFAP, and iNOS were observed in TNF-treated astrocytes. Silencing of β3 Integrin prior to TNF treatment prevented Thy-1-induced migration, while β3 Integrin over-expression was sufficient to induce astrocyte reactivity and allow Thy-1-induced migration. Finally, hSOD1G93A astrocytes behave as TNF-treated astrocytes since they were reactive and responsive to Thy-1. Conclusions Therefore, inflammation induces expression of αVβ3 Integrin and other proteins, astrocyte reactivity, and Thy-1 responsiveness. Importantly, ectopic control of β3 Integrin levels modulates these responses regardless of inflammation. Electronic supplementary material The online version of this article (10.1186/s12974-017-0968-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Raúl Lagos-Cabré
- Cellular Communication Laboratory, Programme of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile.,Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile
| | - Alvaro Alvarez
- Cellular Communication Laboratory, Programme of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile.,Facultad de Ciencia, Universidad San Sebastian, Santiago, Chile
| | - Milene Kong
- Cellular Communication Laboratory, Programme of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile.,Department of Biomedicine, Faculty of Health Sciences, University of Antofagasta, Antofagasta, Chile
| | - Francesca Burgos-Bravo
- Cellular Communication Laboratory, Programme of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile
| | - Areli Cárdenas
- Cellular Communication Laboratory, Programme of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile.,Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile.,Departamento de Ciencias Químicas y Biológicas, Universidad Bernardo O'Higgins, 837-0854, Santiago, Chile
| | - Edgardo Rojas-Mancilla
- Departamento de Ciencias Químicas y Biológicas, Universidad Bernardo O'Higgins, 837-0854, Santiago, Chile
| | - Ramón Pérez-Nuñez
- Cellular Communication Laboratory, Programme of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile.,Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile
| | | | - Fabiola Rojas
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, 1066, Epalinges, Switzerland
| | - Mario Herrera-Marschitz
- Programme of Molecular & Clinical Pharmacology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile
| | - Andrew F G Quest
- Cellular Communication Laboratory, Programme of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile.,Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile
| | - Brigitte van Zundert
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Programme of Cellular & Molecular Biology, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile. .,Center for Molecular Studies of the Cell (CEMC), Advanced Center for Chronic Diseases (ACCDiS), Facultad de Medicina, Universidad de Chile, 838-0453, Santiago, Chile.
| |
Collapse
|
40
|
Allagnat F, Dubuis C, Lambelet M, Le Gal L, Alonso F, Corpataux JM, Déglise S, Haefliger JA. Connexin37 reduces smooth muscle cell proliferation and intimal hyperplasia in a mouse model of carotid artery ligation. Cardiovasc Res 2017; 113:805-816. [PMID: 28449099 DOI: 10.1093/cvr/cvx079] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 04/20/2017] [Indexed: 12/11/2022] Open
Abstract
AIMS Intimal hyperplasia (IH) is an abnormal response to vessel injury characterized by the dedifferentiation, migration, and proliferation of quiescent vascular smooth muscle cells (VSMC) to form a neointima layer. Vascular connexins (Cx) are involved in the pathophysiology of various vascular diseases, and Cx43, the main Cx expressed in VSMC, has been shown to promote VSMC proliferation and IH. The aim of this study was to investigate the participation of another Cx, namely Cx37, in the formation of the neointima layer. METHODS AND RESULTS Wild-type (WT) and Cx37-deficient (Cx37-/-) C57BL/6J mice were subjected to carotid artery ligation (CAL), a model of vessel injury and IH. The neointima developed linearly in WT until 28 days post surgery. In contrast, the neointima layer was almost absent 14 days after surgery in Cx37-/- mice, and twice as more developed after 28 days compared to WT mice. This large neointima formation correlated with a two-fold increase in cell proliferation in the media and neointima regions between 14 and 28 days in Cx37-/- mice compared to WT mice. The CAL triggered Cx43 overexpression in the media and neointima layers of ligated carotids in WT mice, and selectively up-regulated Cx37 expression in the media layer, but not in the neointima layer. The de novo expression of Cx37 in human primary VSMC reduced cell proliferation and P-Akt levels, in association with lower Cx43 levels, whereas Cx43 overexpression increased P-Akt levels. CONCLUSION The presence of Cx37 in the media layer of injured arteries restrains VSMC proliferation and limits the development of IH, presumably by interfering with the pro-proliferative effect of Cx43 and the Akt pathway.
Collapse
MESH Headings
- Aged
- Animals
- Carotid Arteries/metabolism
- Carotid Arteries/pathology
- Carotid Arteries/surgery
- Carotid Artery Injuries/genetics
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/pathology
- Carotid Stenosis/genetics
- Carotid Stenosis/metabolism
- Carotid Stenosis/pathology
- Cell Proliferation
- Cells, Cultured
- Connexin 43/metabolism
- Connexins/deficiency
- Connexins/genetics
- Connexins/metabolism
- Disease Models, Animal
- Female
- Humans
- Hyperplasia
- Ligation
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima
- Phosphorylation
- Proto-Oncogene Proteins c-akt/metabolism
- Signal Transduction
- Time Factors
- Gap Junction alpha-4 Protein
Collapse
Affiliation(s)
- Florent Allagnat
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Céline Dubuis
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Martine Lambelet
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Loïc Le Gal
- Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV), Laboratory of Experimental Medicine, c/o Department of Physiology, Bugnon 7a, 1005 Lausanne, Switzerland
| | - Florian Alonso
- Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV), Laboratory of Experimental Medicine, c/o Department of Physiology, Bugnon 7a, 1005 Lausanne, Switzerland
| | - Jean-Marc Corpataux
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Sébastien Déglise
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Jacques-Antoine Haefliger
- Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV), Laboratory of Experimental Medicine, c/o Department of Physiology, Bugnon 7a, 1005 Lausanne, Switzerland
| |
Collapse
|
41
|
Caliceti C, Rizzo P, Ferrari R, Fortini F, Aquila G, Leoncini E, Zambonin L, Rizzo B, Calabria D, Simoni P, Mirasoli M, Guardigli M, Hrelia S, Roda A, Cicero AFG. Novel role of the nutraceutical bioactive compound berberine in lectin-like OxLDL receptor 1-mediated endothelial dysfunction in comparison to lovastatin. Nutr Metab Cardiovasc Dis 2017; 27:552-563. [PMID: 28511903 DOI: 10.1016/j.numecd.2017.04.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 03/31/2017] [Accepted: 04/10/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND AIMS Oxidized LDL (oxLDL) or pro-inflammatory stimuli lead to increased oxidative stress linked to endothelial dysfunction and atherosclerosis. The oxLDL receptor-1 (LOX1) is elevated within atheromas and cholesterol-lowering statins inhibit LOX1 expression. Berberine (BBR), an alkaloid extracted from plants of gender Berberis, has lipid-lowering and anti-inflammatory activity. However, its role in regulating LOX1-mediated signaling is still unknown. The aim of this study was to investigate the effect of BBR on oxLDL- and TNFα-induced endothelial dysfunction in human umbilical vein endothelial cells (HUVECs) and to compare it with that of lovastatin (LOVA). METHODS AND RESULTS Cytotoxicity was determined by lactate dehydrogenase assay. Antioxidant capacity was measured with chemiluminescent and fluorescent method and intracellular ROS levels through a fluorescent dye. Gene and protein expression levels were assayed by qRT-PCR and western blot, respectively. HUVECs exposure to oxLDL (30 μg/ml) or TNFα (10 ng/ml) for 24 h led to a significant increase in LOX1 expression, effect abrogated by BBR (5 μM) and LOVA (5 μM). BBR but not LOVA treatment abolished the TNFα-induced cytotoxicity and restored the activation of Akt signaling. In spite of a low direct antioxidant capacity, both compounds reduced intracellular ROS levels generated by treatment of TNFα but only BBR inhibited NOX2 expression, MAPK/Erk1/2 signaling and subsequent NF-κB target genes VCAM and ICAM expression, induced by TNFα. CONCLUSIONS These findings demonstrated for the first time that BBR could prevent the oxLDL and TNFα - induced LOX1 expression and oxidative stress, key events that lead to NOX, MAPK/Erk1/2 and NF-κB activation linked to endothelial dysfunction. CHEMICAL COMPOUNDS STUDIED IN THIS ARTICLE Berberine (PubChem CID: 2353); Lovastatin (PubChem CID: 53232).
Collapse
Affiliation(s)
- C Caliceti
- Department of Chemistry "Giacomo Ciamician" - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Centro Interdipartimentale di Ricerca Industriale Energia e Ambiente (CIRI EA) - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Istituto Nazionale Biostrutture e Biosistemi (INBB), Roma, Italy.
| | - P Rizzo
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy; Maria Cecilia Hospital, GVM Care&Research, E.S: Health Science Foundation, Cotignola, Italy
| | - R Ferrari
- Department of Medical Sciences, Cardiology and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy; Maria Cecilia Hospital, GVM Care&Research, E.S: Health Science Foundation, Cotignola, Italy
| | - F Fortini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - G Aquila
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - E Leoncini
- Department for Life Quality Studies - Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - L Zambonin
- Department of Pharmacy and Biotechnology - Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - B Rizzo
- Department for Life Quality Studies - Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - D Calabria
- Centro Interdipartimentale di Ricerca Industriale Energia e Ambiente (CIRI EA) - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Istituto Nazionale Biostrutture e Biosistemi (INBB), Roma, Italy
| | - P Simoni
- Department of Medical and Surgical Sciences-DIMEC, Sant'Orsola Malpighi Hospital, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - M Mirasoli
- Department of Chemistry "Giacomo Ciamician" - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Centro Interdipartimentale di Ricerca Industriale Energia e Ambiente (CIRI EA) - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Istituto Nazionale Biostrutture e Biosistemi (INBB), Roma, Italy
| | - M Guardigli
- Department of Chemistry "Giacomo Ciamician" - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Istituto Nazionale Biostrutture e Biosistemi (INBB), Roma, Italy
| | - S Hrelia
- Department for Life Quality Studies - Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - A Roda
- Department of Chemistry "Giacomo Ciamician" - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Centro Interdipartimentale di Ricerca Industriale Energia e Ambiente (CIRI EA) - Alma Mater Studiorum, University of Bologna, Bologna, Italy; Istituto Nazionale Biostrutture e Biosistemi (INBB), Roma, Italy
| | - A F G Cicero
- Department of Medical and Surgical Sciences-DIMEC, Sant'Orsola Malpighi Hospital, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| |
Collapse
|
42
|
Li H, Chai S, Dai L, Gu C. Collagen External Scaffolds Mitigate Intimal Hyperplasia and Improve Remodeling of Vein Grafts in a Rabbit Arteriovenous Graft Model. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7473437. [PMID: 28503573 PMCID: PMC5414509 DOI: 10.1155/2017/7473437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/13/2017] [Accepted: 03/26/2017] [Indexed: 11/18/2022]
Abstract
Objectives. The aim of this study was to test the effects of collagen external scaffold (CES) in intimal hyperplasia of vein grafts and explore its underlying mechanisms. Methods. Thirty-six New Zealand white rabbits were randomized into no-graft group, graft group, and CES group. The rabbit arteriovenous graft model was established. In CES group, the vein graft was wrapped around with CES. The hemodynamic parameters of vein grafts were measured intraoperatively and 4 weeks after operation by ultrasonic examination. Histological characteristics of vein grafts were also evaluated 4 weeks later. The mRNA and protein levels of proliferating cell nuclear antigen (PCNA), active cleaved-caspase-3 (ClvCasp-3), and smooth muscle 22 alpha (SM22α) were measured 4 weeks later by quantitative real-time PCR and western blot. Results. CES significantly improved the hemodynamic stability of vein grafts, with higher blood velocity and blood flow. Similarly, CES also markedly mitigated intimal hyperplasia and inhibited dilatation of vein grafts. In CES group, the upexpression of PCNA and ClvCasp-3 and the downexpression of SM22α were inhibited. Conclusion. CES exerts beneficial effects in mitigating intimal hyperplasia and improving remodeling of autogenous vein grafts, which may be associated with reducing the proliferation and apoptosis and preserving the phenotype of VSMCs.
Collapse
Affiliation(s)
- Haiming Li
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Shoudong Chai
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Longsheng Dai
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chengxiong Gu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
43
|
Meda P. Gap junction proteins are key drivers of endocrine function. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:124-140. [PMID: 28284720 DOI: 10.1016/j.bbamem.2017.03.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 01/07/2023]
Abstract
It has long been known that the main secretory cells of exocrine and endocrine glands are connected by gap junctions, made by a variety of connexin species that ensure their electrical and metabolic coupling. Experiments in culture systems and animal models have since provided increasing evidence that connexin signaling contributes to control the biosynthesis and release of secretory products, as well as to the life and death of secretory cells. More recently, genetic studies have further provided the first lines of evidence that connexins also control the function of human glands, which are central to the pathogenesis of major endocrine diseases. Here, we summarize the recent information gathered on connexin signaling in these systems, since the last reviews on the topic, with particular regard to the pancreatic beta cells which produce insulin, and the renal cells which produce renin. These cells are keys to the development of various forms of diabetes and hypertension, respectively, and combine to account for the exploding, worldwide prevalence of the metabolic syndrome. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, Switzerland.
| |
Collapse
|
44
|
Li H, Spagnol G, Zheng L, Stauch KL, Sorgen PL. Regulation of Connexin43 Function and Expression by Tyrosine Kinase 2. J Biol Chem 2016; 291:15867-80. [PMID: 27235399 DOI: 10.1074/jbc.m116.727008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Indexed: 11/06/2022] Open
Abstract
Connexin43 (Cx43) assembly and degradation, the regulation of electrical and metabolic coupling, as well as modulating the interaction with other proteins, involve phosphorylation. Here, we identified and characterized the biological significance of a novel tyrosine kinase that phosphorylates Cx43, tyrosine kinase 2 (Tyk2). Activation of Tyk2 led to a decrease in Cx43 gap junction communication by increasing the turnover rate of Cx43 from the plasma membrane. Tyk2 directly phosphorylated Cx43 residues Tyr-247 and Tyr-265, leading to indirect phosphorylation on residues Ser-279/Ser-282 (MAPK) and Ser-368 (PKC). Although this phosphorylation pattern is similar to what has been observed following Src activation, the response caused by Tyk2 occurred when Src was inactive in NRK cells. Knockdown of Tyk2 at the permissive temperature (active v-Src) in LA-25 cells decreased Cx43 phosphorylation, indicating that although activation of Tyk2 and v-Src leads to phosphorylation of the same Cx43CT residues, they are not identical in level at each site. Additionally, angiotensin II activation of Tyk2 increased the intracellular protein level of Cx43 via STAT3. These findings indicate that, like Src, Tyk2 can also inhibit gap junction communication by phosphorylating Cx43.
Collapse
Affiliation(s)
- Hanjun Li
- From the University of Nebraska Medical Center, Omaha, Nebraska 68105
| | - Gaelle Spagnol
- From the University of Nebraska Medical Center, Omaha, Nebraska 68105
| | - Li Zheng
- From the University of Nebraska Medical Center, Omaha, Nebraska 68105
| | - Kelly L Stauch
- From the University of Nebraska Medical Center, Omaha, Nebraska 68105
| | - Paul L Sorgen
- From the University of Nebraska Medical Center, Omaha, Nebraska 68105
| |
Collapse
|
45
|
Nitric Oxide Deficit Drives Intimal Hyperplasia in Mouse Models of Hypertension. Eur J Vasc Endovasc Surg 2016; 51:733-42. [DOI: 10.1016/j.ejvs.2016.01.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 01/29/2016] [Indexed: 01/26/2023]
|
46
|
Munger SJ, Geng X, Srinivasan RS, Witte MH, Paul DL, Simon AM. Segregated Foxc2, NFATc1 and Connexin expression at normal developing venous valves, and Connexin-specific differences in the valve phenotypes of Cx37, Cx43, and Cx47 knockout mice. Dev Biol 2016; 412:173-90. [PMID: 26953188 PMCID: PMC4826804 DOI: 10.1016/j.ydbio.2016.02.033] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 02/12/2016] [Accepted: 02/12/2016] [Indexed: 01/25/2023]
Abstract
Venous valves (VVs) are critical for unidirectional blood flow from superficial and deep veins towards the heart. Congenital valve aplasia or agenesis may, in some cases, be a direct cause of vascular disease, motivating an understanding of the molecular mechanisms underlying the development and maintenance of VVs. Three gap junction proteins (Connexins), Cx37, Cx43, and Cx47, are specifically expressed at VVs in a highly polarized fashion. VVs are absent from adult mice lacking Cx37; however it is not known if Cx37 is required for the initial formation of valves. In addition, the requirement of Cx43 and Cx47 for VV development has not been studied. Here, we provide a detailed description of Cx37, Cx43, and Cx47 expression during mouse vein development and show by gene knockout that each Cx is necessary for normal valve development. The valve phenotypes in the knockout lines exhibit Cx-specific differences, however, including whether peripheral or central VVs are affected by gene inactivation. In addition, we show that a Cx47 null mutation impairs peripheral VV development but does not affect lymphatic valve formation, a finding of significance for understanding how some CX47 mutations cause inherited lymphedema in humans. Finally, we demonstrate a striking segregation of Foxc2 and NFATc1 transcription factor expression between the downstream and upstream faces, respectively, of developing VV leaflets and show that this segregation is closely associated with the highly polarized expression of Cx37, Cx43, and Cx47. The partition of Foxc2 and NFATc1 expression at VV leaflets makes it unlikely that these factors directly cooperate during the leaflet elongation stage of VV development.
Collapse
Affiliation(s)
| | - Xin Geng
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
| | - Marlys H Witte
- Department of Surgery, University of Arizona, Tucson, AZ 85724, USA.
| | - David L Paul
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| | - Alexander M Simon
- Department of Physiology, University of Arizona, Tucson, AZ 85724, USA.
| |
Collapse
|
47
|
Alonso F, Domingos-Pereira S, Le Gal L, Derré L, Meda P, Jichlinski P, Nardelli-Haefliger D, Haefliger JA. Targeting endothelial connexin40 inhibits tumor growth by reducing angiogenesis and improving vessel perfusion. Oncotarget 2016; 7:14015-28. [PMID: 26883111 PMCID: PMC4924695 DOI: 10.18632/oncotarget.7370] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 01/29/2016] [Indexed: 01/09/2023] Open
Abstract
Endothelial connexin40 (Cx40) contributes to regulate the structure and function of vessels. We have examined whether the protein also modulates the altered growth of vessels in tumor models established in control mice (WT), mice lacking Cx40 (Cx40-/-), and mice expressing the protein solely in endothelial cells (Tie2-Cx40). Tumoral angiogenesis and growth were reduced, whereas vessel perfusion, smooth muscle cell (SMC) coverage and animal survival were increased in Cx40-/- but not Tie2-Cx40 mice, revealing a critical involvement of endothelial Cx40 in transformed tissues independently of the hypertensive status of Cx40-/- mice. As a result, Cx40-/- mice bearing tumors survived significantly longer than corresponding controls, including after a cytotoxic administration. Comparable observations were made in WT mice injected with a peptide targeting Cx40, supporting the Cx40 involvement. This involvement was further confirmed in the absence of Cx40 or by peptide-inhibition of this connexin in aorta-sprouting, matrigel plug and SMC migration assays, and associated with a decreased expression of the phosphorylated form of endothelial nitric oxide synthase. The data identify Cx40 as a potential novel target in cancer treatment.
Collapse
MESH Headings
- Animals
- Aorta/pathology
- Apoptosis
- Biomarkers, Tumor/metabolism
- Blood Vessels/physiology
- Cell Proliferation
- Connexins/antagonists & inhibitors
- Connexins/metabolism
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Female
- Humans
- Lung Neoplasms/blood supply
- Lung Neoplasms/pathology
- Lung Neoplasms/prevention & control
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/pathology
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasm Invasiveness
- Neovascularization, Pathologic/prevention & control
- Perfusion
- Tumor Cells, Cultured
- Urinary Bladder Neoplasms/blood supply
- Urinary Bladder Neoplasms/pathology
- Urinary Bladder Neoplasms/prevention & control
- Gap Junction alpha-5 Protein
Collapse
Affiliation(s)
- Florian Alonso
- Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | | | - Loïc Le Gal
- Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Laurent Derré
- Department of Urology, Lausanne University Hospital, Lausanne, Switzerland
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva, Medical Center, Geneva, Switzerland
| | - Patrice Jichlinski
- Department of Urology, Lausanne University Hospital, Lausanne, Switzerland
| | | | | |
Collapse
|
48
|
Longchamp A, Allagnat F, Alonso F, Kuppler C, Dubuis C, Ozaki CK, Mitchell JR, Berceli S, Corpataux JM, Déglise S, Haefliger JA. Connexin43 Inhibition Prevents Human Vein Grafts Intimal Hyperplasia. PLoS One 2015; 10:e0138847. [PMID: 26398895 PMCID: PMC4580578 DOI: 10.1371/journal.pone.0138847] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 09/04/2015] [Indexed: 12/12/2022] Open
Abstract
Venous bypass grafts often fail following arterial implantation due to excessive smooth muscle cells (VSMC) proliferation and consequent intimal hyperplasia (IH). Intercellular communication mediated by Connexins (Cx) regulates differentiation, growth and proliferation in various cell types. Microarray analysis of vein grafts in a model of bilateral rabbit jugular vein graft revealed Cx43 as an early upregulated gene. Additional experiments conducted using an ex-vivo human saphenous veins perfusion system (EVPS) confirmed that Cx43 was rapidly increased in human veins subjected ex-vivo to arterial hemodynamics. Cx43 knock-down by RNA interference, or adenoviral-mediated overexpression, respectively inhibited or stimulated the proliferation of primary human VSMC in vitro. Furthermore, Cx blockade with carbenoxolone or the specific Cx43 inhibitory peptide 43gap26 prevented the burst in myointimal proliferation and IH formation in human saphenous veins. Our data demonstrated that Cx43 controls proliferation and the formation of IH after arterial engraftment.
Collapse
Affiliation(s)
- Alban Longchamp
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois, Laboratory of Experimental Medicine, Lausanne, Switzerland
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Florent Allagnat
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois, Laboratory of Experimental Medicine, Lausanne, Switzerland
| | - Florian Alonso
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois, Laboratory of Experimental Medicine, Lausanne, Switzerland
| | - Christopher Kuppler
- Malcom Randall Veterans Affairs Medical Center and the Division of Vascular and Endovascular Surgery, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Céline Dubuis
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois, Laboratory of Experimental Medicine, Lausanne, Switzerland
| | - Charles-Keith Ozaki
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - James R. Mitchell
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts, United States of America
| | - Scott Berceli
- Malcom Randall Veterans Affairs Medical Center and the Division of Vascular and Endovascular Surgery, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Jean-Marc Corpataux
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois, Laboratory of Experimental Medicine, Lausanne, Switzerland
| | - Sébastien Déglise
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois, Laboratory of Experimental Medicine, Lausanne, Switzerland
| | - Jacques-Antoine Haefliger
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois, Laboratory of Experimental Medicine, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
49
|
Zhang J, Yang GM, Zhu Y, Peng XY, Li T, Liu LM. Role of connexin 43 in vascular hyperpermeability and relationship to Rock1-MLC20 pathway in septic rats. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1323-32. [PMID: 26342084 DOI: 10.1152/ajplung.00016.2015] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 08/31/2015] [Indexed: 12/27/2022] Open
Abstract
Connexin (Cx)43 has been shown to participate in several cardiovascular diseases. Increased vascular permeability is a common and severe complication in sepsis or septic shock. Whether or not Cx43 takes part in the regulation of vascular permeability in severe sepsis is not known, and the underlying mechanism has not been described. With cecal ligation and puncture-induced sepsis in rats and lipopolysaccharide (LPS)-treated vascular endothelial cells (VECs) from pulmonary veins, the role of Cx43 in increased vascular permeability and its relationship to the RhoA/Rock1 pathway were studied. It was shown that vascular permeability in the lungs, kidneys, and mesentery in sepsis rats and LPS-stimulated monolayer pulmonary vein VECs was significantly increased and positively correlated with the increased expression of Cx43 and Rock1 in these organs and cultured pulmonary vein VECs. The connexin inhibitor carbenoxolone (10 mg/kg iv) and the Rock1 inhibitor Y-27632 (2 mg/kg iv) alleviated the vascular leakage of lung, mesentery, and kidney in sepsis rats. Overexpressed Cx43 increased the phosphorylation of 20-kDa myosin light chain (MLC20) and the expression of Rock1 and increased the vascular permeability and decreased the transendothelial electrical resistance of pulmonary vein VECs. Cx43 RNA interference decreased the phosphorylation of MLC20 and the expression of Rock1 and decreased LPS-stimulated hyperpermeability of cultured pulmonary vein VECs. The Rock1 inhibitor Y-27632 alleviated LPS- and overexpressed Cx43-induced hyperpermeability of monolayer pulmonary vein VECs. This report shows that Cx43 participates in the regulation of vascular permeability in sepsis and that the mechanism is related to the Rock1-MLC20 phosphorylation pathway.
Collapse
Affiliation(s)
- Jie Zhang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Guang-Ming Yang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Yu Zhu
- State Key Laboratory of Trauma, Burns, and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Xiao-Yong Peng
- State Key Laboratory of Trauma, Burns, and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Tao Li
- State Key Laboratory of Trauma, Burns, and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Liang-Ming Liu
- State Key Laboratory of Trauma, Burns, and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People's Republic of China
| |
Collapse
|
50
|
Abed AB, Kavvadas P, Chadjichristos CE. Functional roles of connexins and pannexins in the kidney. Cell Mol Life Sci 2015; 72:2869-77. [PMID: 26082183 PMCID: PMC11113829 DOI: 10.1007/s00018-015-1964-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 12/22/2022]
Abstract
Kidneys are highly complex organs, playing a crucial role in human physiopathology, as they are implicated in vital processes, such as fluid filtration and vasomotor tone regulation. There is growing evidence that gap junctions are major determinants of renal physiopathology. It has been demonstrated that their expression or channel activity may vary depending on physiological and pathological situations within distinct renal compartments. While some studies have focused on the role of connexins in renal physiology, our knowledge regarding the functional relevance of pannexins is still very limited. In this paper, we provide an overview of the involvement of connexins, pannexins and their channels in various physiological processes related to different renal compartments.
Collapse
Affiliation(s)
- Ahmed B. Abed
- INSERM UMR-S1155, Batiment Recherche, Tenon Hospital, 4 rue de la Chine, 75020 Paris, France
- Sorbonne Universite´s, UPMC Univ Paris 6, Paris, France
| | - Panagiotis Kavvadas
- INSERM UMR-S1155, Batiment Recherche, Tenon Hospital, 4 rue de la Chine, 75020 Paris, France
| | | |
Collapse
|