1
|
Capalbo S, Polyakova A, El Imane Z, Khan I, Kawai T, Shindo S, Salinas M. A Comprehensive Review of Contemporary Bioreactors for Vascular Inflammation Studies. Inflammation 2025:10.1007/s10753-024-02231-y. [PMID: 39903422 DOI: 10.1007/s10753-024-02231-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 12/11/2024] [Accepted: 12/26/2024] [Indexed: 02/06/2025]
Abstract
The field of vascular biology has advanced significantly with bioreactor systems, which have become essential tools for investigating the mechanisms of vascular inflammatory diseases such as atherosclerosis, vasculitis, and aneurysms. These bioreactors allow researchers to recreate specific vascular environments, providing a controlled setting for studying the effects of blood flow, mechanical stress, and biochemical factors on vascular tissues. Through these systems, researchers can explore how physical and chemical cues contribute to disease processes and cellular responses, enhancing our understanding of disease progression. Bioreactor studies have demonstrated that hemodynamic forces, particularly shear stress, influence endothelial cell behavior and play a role in vascular pathologies. For instance, in atherosclerosis, disturbed flow patterns are associated with endothelial dysfunction and plaque development. By simulating these conditions, bioreactors provide insight into the effects of mechanical forces on vascular wall biology, highlighting how altered flow can contribute to disease. Bioreactors also support studies on the impacts of pulsatile flow and circumferential stress, allowing a closer approximation of physiological environments. Beyond flow dynamics, these systems facilitate investigation into how vascular cells respond to biochemical signals, inflammatory markers, and therapeutic interventions. This integrated approach allows for a more complete picture of the factors involved in vascular disease. Recent advancements, such as vessel-on-a-chip models and artery-mimicking setups, extend the capabilities of bioreactors by enabling researchers to model a broader range of conditions relevant to human physiology. In vasculitis studies, bioreactors help explore immune interactions with endothelial cells, especially with stem cell-derived cells that replicate patient-specific responses. Bioreactors also play a role in vascular tissue engineering, particularly in assessing materials and scaffold-free designs that may reduce inflammation in vascular grafts. These efforts contribute to the ongoing search for more compatible graft materials, with the potential to improve outcomes in clinical applications. This review provides a comprehensive overview of bioreactor technologies applied in vascular inflammation research, examining their designs, applications, and contributions to disease modeling. Organized into sections on bioreactor configurations, flow dynamics, biochemical interactions, and tissue engineering applications, the review concludes by discussing recent innovations and highlighting directions for future research, underscoring the role of bioreactors in bridging laboratory studies with insights into vascular disease.
Collapse
Affiliation(s)
- Solana Capalbo
- Nova Southeastern University, College of Computing and Engineering, Davie, FL, USA
| | | | - Zayd El Imane
- Nova Southeastern University, College of Computing and Engineering, Davie, FL, USA
| | - Izza Khan
- Nova Southeastern University, College of Computing and Engineering, Davie, FL, USA
| | - Toshihisa Kawai
- Department of Oral Science and Translational Research, Nova Southeastern University College of Dental Medicine, Fort Lauderdale, FL, USA
| | - Satoru Shindo
- Department of Oral Science and Translational Research, Nova Southeastern University College of Dental Medicine, Fort Lauderdale, FL, USA
| | - Manuel Salinas
- Nova Southeastern University, College of Computing and Engineering, Davie, FL, USA.
| |
Collapse
|
2
|
Peng S, Wu WQ, Li LY, Shi YC, Lin S, Song ZY. Deficiency of neuropeptide Y attenuates neointima formation after vascular injury in mice. BMC Cardiovasc Disord 2023; 23:239. [PMID: 37149580 PMCID: PMC10164319 DOI: 10.1186/s12872-023-03267-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 04/26/2023] [Indexed: 05/08/2023] Open
Abstract
BACKGROUND Restenosis after percutaneous coronary intervention (PCI) limits therapeutic revascularization. Neuropeptide Y (NPY), co-stored and co-released with the sympathetic nervous system, is involved in this process, but its exact role and underlying mechanisms remain to be fully understood. This study aimed to investigate the role of NPY in neointima formation after vascular injury. METHODS Using the left carotid arteries of wild-type (WT, NPY-intact) and NPY-deficient (NPY-/-) mice, ferric chloride-mediated carotid artery injury induced neointima formation. Three weeks after injury, the left injured carotid artery and contralateral uninjured carotid artery were collected for histological analysis and immunohistochemical staining. RT-qPCR was used to detect the mRNA expression of several key inflammatory markers and cell adhesion molecules in vascular samples. Raw264.7 cells were treated with NPY, lipopolysaccharide (LPS), and lipopolysaccharide-free, respectively, and RT-qPCR was used to detect the expression of these inflammatory mediators. RESULTS Compared with WT mice, NPY-/- mice had significantly reduced neointimal formation three weeks after injury. Mechanistically, immunohistochemical analysis showed there were fewer macrophages and more vascular smooth muscle cells in the neointima of NPY-/- mice. Moreover, the mRNA expression of key inflammatory markers such as interleukin-6 (IL-6), transforming growth factor-β1 (TGF-β1), and intercellular adhesion molecule-1 (ICAM-1) was significantly lower in the injured carotid arteries of NPY-/- mice, compared to that in the injured carotid arteries of WT mice. In RAW264.7 macrophages, NPY significantly promoted TGF-β1 mRNA expression under unactivated but not LPS-stimulated condition. CONCLUSIONS Deletion of NPY attenuated neointima formation after artery injury, at least partly, through reducing the local inflammatory response, suggesting that NPY pathway may provide new insights into the mechanism of restenosis.
Collapse
Affiliation(s)
- Song Peng
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wei-Qiang Wu
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lin-Yu Li
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yan-Chuan Shi
- Group of Neuroendocrinology, Garvan Institute of Medical Research, 384 Victoria St, Sydney, Australia
| | - Shu Lin
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
- Group of Neuroendocrinology, Garvan Institute of Medical Research, 384 Victoria St, Sydney, Australia.
| | - Zhi-Yuan Song
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
3
|
Liu S, Lin Z. Vascular Smooth Muscle Cells Mechanosensitive Regulators and Vascular Remodeling. J Vasc Res 2021; 59:90-113. [PMID: 34937033 DOI: 10.1159/000519845] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 09/23/2021] [Indexed: 11/19/2022] Open
Abstract
Blood vessels are subjected to mechanical loads of pressure and flow, inducing smooth muscle circumferential and endothelial shear stresses. The perception and response of vascular tissue and living cells to these stresses and the microenvironment they are exposed to are critical to their function and survival. These mechanical stimuli not only cause morphological changes in cells and vessel walls but also can interfere with biochemical homeostasis, leading to vascular remodeling and dysfunction. However, the mechanisms underlying how these stimuli affect tissue and cellular function, including mechanical stimulation-induced biochemical signaling and mechanical transduction that relies on cytoskeletal integrity, are unclear. This review focuses on signaling pathways that regulate multiple biochemical processes in vascular mesangial smooth muscle cells in response to circumferential stress and are involved in mechanosensitive regulatory molecules in response to mechanotransduction, including ion channels, membrane receptors, integrins, cytoskeletal proteins, nuclear structures, and cascades. Mechanoactivation of these signaling pathways is closely associated with vascular remodeling in physiological or pathophysiological states.
Collapse
Affiliation(s)
- Shangmin Liu
- Ji Hua Institute of Biomedical Engineering Technology, Ji Hua Laboratory, Foshan, China, .,Medical Research Center, Guangdong Academy of Medical Sciences, Guangdong General Hospital, Guangzhou, China,
| | - Zhanyi Lin
- Ji Hua Institute of Biomedical Engineering Technology, Ji Hua Laboratory, Foshan, China.,Institute of Geriatric Medicine, Guangdong Academy of Medical Sciences, Guangdong General Hospital, Guangzhou, China
| |
Collapse
|
4
|
Cell signaling model for arterial mechanobiology. PLoS Comput Biol 2020; 16:e1008161. [PMID: 32834001 PMCID: PMC7470387 DOI: 10.1371/journal.pcbi.1008161] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 09/03/2020] [Accepted: 07/17/2020] [Indexed: 11/20/2022] Open
Abstract
Arterial growth and remodeling at the tissue level is driven by mechanobiological processes at cellular and sub-cellular levels. Although it is widely accepted that cells seek to promote tissue homeostasis in response to biochemical and biomechanical cues—such as increased wall stress in hypertension—the ways by which these cues translate into tissue maintenance, adaptation, or maladaptation are far from understood. In this paper, we present a logic-based computational model for cell signaling within the arterial wall, aiming to predict changes in extracellular matrix turnover and cell phenotype in response to pressure-induced wall stress, flow-induced wall shear stress, and exogenous sources of angiotensin II, with particular interest in mouse models of hypertension. We simulate a number of experiments from the literature at both the cell and tissue level, involving single or combined inputs, and achieve high qualitative agreement in most cases. Additionally, we demonstrate the utility of this modeling approach for simulating alterations (in this case knockdowns) of individual nodes within the signaling network. Continued modeling of cellular signaling will enable improved mechanistic understanding of arterial growth and remodeling in health and disease, and will be crucial when considering potential pharmacological interventions. Biological soft tissues are characterized by continuous production and removal of material, which endows them with a remarkable ability to adapt to changes in their biochemical and biomechanical environments. For arteries, mechanical stimuli result primarily from changes in blood pressure or flow, and biochemical changes are induced by multiple factors, including pharmacological intervention. In order to understand how arterial properties are maintained in health, or how they adapt or fail to adapt in disease, we must understand better how these diverse stimuli affect material turnover. Extracellular matrix is tightly regulated by mechano-sensing and mechano-regulation, and therefore cell signaling, thus we present a computational model of relevant signaling pathways within the vascular wall, with the aim of predicting changes in wall composition and function in response to three main inputs: pressure-induced wall stress, flow-induced wall shear stress, and exogenous angiotensin II. We obtain qualitative agreement with a range of experimental studies from the literature, and provide illustrative examples demonstrating how such models can be used to further our understanding of arterial remodeling.
Collapse
|
5
|
Wang L, Deng L, Lin N, Shi Y, Chen J, Zhou Y, Chen D, Liu S, Li C. Berberine inhibits proliferation and apoptosis of vascular smooth muscle cells induced by mechanical stretch via the PDI/ERS and MAPK pathways. Life Sci 2020; 259:118253. [PMID: 32795536 DOI: 10.1016/j.lfs.2020.118253] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/03/2020] [Accepted: 08/08/2020] [Indexed: 12/13/2022]
Abstract
AIMS We recently demonstrated that mechanical stretch increases the proliferation and apoptosis of vascular smooth muscle cells (VSMCs) by activating the protein disulfide isomerase (PDI) redox system, thus accelerating atherosclerotic lesion formation in the transplanted vein. At present, there are no efficient intervention measures to prevent this phenomenon. Berberine inhibits pathological vascular remodeling caused by hypertension, but the underlying mechanism is controversial. Herein, we investigate the role of berberine and the underlying mechanism of its effects on mechanical stretch-induced VSMC proliferation and apoptosis. MAIN METHODS Mouse VSMCs cultivated on flexible membranes were pretreated for 1 h with one of the following substances: berberine, PDI inhibitor bacitracin, MAPK inhibitors, or ERS inhibitor 4-PBA. VSMCs were then subjected to mechanical stretch. Immunofluorescence and western blot were used to detect proliferation and apoptosis, as well as to analyze signaling pathways in VSMCs. KEY FINDINGS Our results showed that berberine inhibits the PDI-endoplasmic reticulum stress system, thereby attenuating the simultaneous increase of VSMC proliferation and apoptosis in response to mechanical stretch. Interestingly, MAPK inhibitors PD98059, SP600125, and SB202190 significantly reduced the activation of ERS signaling cascades, and their combination with berberine had additive effects. The ERS inhibitor 4-PBA reduced PDI activation and ERS signaling, but not MAPK phosphorylation. Moreover, caspase-3 and caspase-12 were downregulated by berberine. SIGNIFICANCE These results illustrate a novel mechanism of action of berberine that has practical implications. Our data provide important insights for the prevention and treatment of vascular remodeling and diseases caused by mechanical stretching during hypertension.
Collapse
Affiliation(s)
- Linli Wang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Lie Deng
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Ning Lin
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Yi Shi
- Division of Vascular Surgery, The First Affiliated Hospital of Sun Yat-sen University, China
| | - Jingbo Chen
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Yan Zhou
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Dadi Chen
- Experimental Center for Basic Medical Teaching, Zhongshan School of Medicine, Sun Yat-sen University, China
| | - Shuying Liu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China.
| | - Chaohong Li
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, China.
| |
Collapse
|
6
|
Chen J, Zhou Y, Liu S, Li C. Biomechanical signal communication in vascular smooth muscle cells. J Cell Commun Signal 2020; 14:357-376. [PMID: 32780323 DOI: 10.1007/s12079-020-00576-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
Biomechanical stresses are closely associated with cardiovascular development and diseases. In vivo, vascular smooth muscle cells are constantly stimulated by biomechanical factors caused by increased blood pressure leading to the non-specific activation of cell transmembrane proteins. Thus, various intracellular signal molecules are simultaneously activated via signaling cascades, which are closely related to alterations in the differentiation, phenotype, inflammation, migration, pyroptosis, calcification, proliferation, and apoptosis of vascular smooth muscle cells. Meanwhile, mechanical stress-induced miRNAs and epigenetics modification on vascular smooth muscle cells play critical roles as well. Eventually, the overall pathophysiology of the cells is altered, resulting in the development of many major clinical diseases, including hypertension, atherosclerosis, grafted venous atherosclerosis, and aneurysm, among others. In this paper, important advances in mechanical signal communication in vascular smooth muscle cells are reviewed.
Collapse
Affiliation(s)
- Jingbo Chen
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan Zhou
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuying Liu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Chaohong Li
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
7
|
Onal EM, Afsar B, Covic A, Vaziri ND, Kanbay M. Gut microbiota and inflammation in chronic kidney disease and their roles in the development of cardiovascular disease. Hypertens Res 2018; 42:123-140. [PMID: 30504819 DOI: 10.1038/s41440-018-0144-z] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 07/25/2018] [Accepted: 07/26/2018] [Indexed: 02/06/2023]
Abstract
The health and proper functioning of the cardiovascular and renal systems largely depend on crosstalk in the gut-kidney-heart/vessel triangle. Recent evidence suggests that the gut microbiota has an integral function in this crosstalk. Mounting evidence indicates that the development of chronic kidney and cardiovascular diseases follows chronic inflammatory processes that are affected by the gut microbiota via various immune, metabolic, endocrine, and neurologic pathways. Additionally, deterioration of the function of the cardiovascular and renal systems has been reported to disrupt the original gut microbiota composition, further contributing to the advancement of chronic cardiovascular and renal diseases. Considering the interaction between the gut microbiota and the renal and cardiovascular systems, we can infer that interventions for the gut microbiota through diet and possibly some medications can prevent/stop the vicious cycle between the gut microbiota and the cardiovascular/renal systems, leading to a decrease in chronic cardiovascular and renal diseases.
Collapse
Affiliation(s)
- Emine M Onal
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Baris Afsar
- Department of Medicine, Division of Nephrology, Suleyman Demirel University School of Medicine, Isparta, Turkey
| | - Adrian Covic
- Nephrology Clinic, Dialysis and Renal Transplant Center, 'C.I. PARHON' University Hospital, and 'Grigore T. Popa' University of Medicine, Iasi, Romania
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, Schools of Medicine and Biological Science, University of California, California, CA, USA
| | - Mehmet Kanbay
- Department of Medicine, Division of Nephrology, Koc University School of Medicine, Istanbul, Turkey.
| |
Collapse
|
8
|
Zhang Z, Zhang D, Du B, Chen Z. Hyperoside inhibits the effects induced by oxidized low-density lipoprotein in vascular smooth muscle cells via oxLDL-LOX-1-ERK pathway. Mol Cell Biochem 2017; 433:169-176. [PMID: 28434118 PMCID: PMC5554480 DOI: 10.1007/s11010-017-3025-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 04/01/2017] [Indexed: 01/05/2023]
Abstract
Hyperoside is a major active constituent in many medicinal plants traditionally used in Chinese medicines for their anti-inflammatory, antioxidative, and vascular protective effects. Recent studies have focused on the protective effects of hyperoside on hyperlipidemia. However, the molecular mechanisms underlying these effects are unknown. In this study, vascular smooth muscle cells (VSMCs) were treated in vitro with oxidized low-density lipoprotein (oxLDL) in the presence or absence of hyperoside. Western blotting, quantitative PCR, and tetrazolium assay were used to detect lectin-like oxLDL receptor-1 (LOX-1) expression and extracellular signal-regulated kinases (ERK) activation, and to determine VSMCs viability. The results demonstrated that oxLDL promoted LOX-1 expression, ERK activation, and proliferation in VSMCs. Hyperoside significantly inhibited the oxLDL-stimulated effects after long time exposure. However, it promoted ERK activation directly following a short incubation duration (25 min). In conclusion, hyperoside inhibits oxLDL-induced LOX-1 expression, ERK activation, and cell proliferation through the oxLDL-LOX-1-ERK pathway in VSMCs. Our findings suggest a novel role of hyperoside in treating and preventing atherosclerosis.
Collapse
Affiliation(s)
- Zhengyu Zhang
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China. .,Department of Cardiology, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Dongdong Zhang
- Department of Ultrasonic, Liaocheng People's Hospital, Liaocheng, 252000, China
| | - Baoling Du
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.,Department of Cardiology, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhiqiang Chen
- Department of Orthopedics, Liaocheng People's Hospital, Liaocheng, 252000, China
| |
Collapse
|
9
|
Wang J, Liu K, Wang H, Li Z, Li Y, Ping S, Bardeesi ASA, Guo Y, Zhou Y, Pei T, Deng L, Sheng P, Liu S, Li C. Role of nifedipine and hydrochlorothiazide in MAPK activation and vascular smooth muscle cell proliferation and apoptosis. Herz 2016; 42:573-584. [DOI: 10.1007/s00059-016-4489-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 08/28/2016] [Accepted: 09/25/2016] [Indexed: 10/20/2022]
|
10
|
Strain induced esophageal growth in a novel rodent model. J Pediatr Surg 2016; 51:1273-8. [PMID: 26976775 DOI: 10.1016/j.jpedsurg.2016.01.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 01/09/2016] [Accepted: 01/11/2016] [Indexed: 10/22/2022]
Abstract
PURPOSE Longitudinal esophageal strain has been shown to increase esophageal length but the contribution of tissue hyperplasia to this growth is unknown. We used a novel model of esophageal stretch to determine the cellular response to the strain stimulus. METHODS Male Sprague-Dawley rats underwent transection of the distal esophagus. The distal stump was ligated and stretched over a silicone tube. The proximal esophageal stump was anastomosed to the stomach to restore continuity. After two, four, or seven days, the silicone tube was removed and the esophageal segment was measured and compared to its initial length. Sham animals had only a thin piece of silicone tubing placed. Standardized histologic sections were evaluated for wall thickness. Immunofluorescence with DAPI, Ki-67, and Myogenin antibodies was used to assess nuclear density, proliferation indices, and myoblast differentiation indices. RESULTS Experimental animals demonstrated a significant increase in esophageal length compared to sham controls at four and seven days with no difference at two days. There was significant lengthening between four and seven days among the experimental animals. There was no change in wall thickness between experimental and sham animals at any time point. Nuclear density was increased at all time points, although this only reached significance at day four. Proliferation indices were significantly increased relative to sham controls at all time points. Esophageal strain induced significantly increased myoblast differentiation. CONCLUSION In this novel rat model of esophageal strain, lengthening is associated with stable esophageal wall thickness, increased nuclear density, increased cellular proliferation, and increased myogenin expression. These data suggest that true tissue hyperplasia may contribute to the increased length seen after esophageal strain.
Collapse
|
11
|
Bo Z, Bin G, Jing W, Cuifang W, Liping A, Jinglin M, Jin J, Xiaoyi T, Cong C, Ning D, Yayi X. Fluid shear stress promotes osteoblast proliferation via the Gαq-ERK5 signaling pathway. Connect Tissue Res 2016; 57:299-306. [PMID: 27115838 DOI: 10.1080/03008207.2016.1181063] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Fluid shear stress (FSS) is a ubiquitous mechanical stimulus that potently promotes osteoblast proliferation. Previously, we reported that extracellular signal-regulated kinase 5 (ERK5) is essential for FSS-induced osteoblast proliferation. However, the precise mechanism by which FSS promotes osteoblast proliferation via ERK5 activation is poorly understood. The aim of this study was to determine the critical role of Gαq in FSS-induced ERK5 phosphorylation and osteoblast proliferation, as well as the downstream targets of the Gαq-ERK5 pathway. MC3T3-E1 cells were transfected with 50 nM Gαq siRNA, treated with 5 mM XMD8-92 (a highly selective inhibitor of ERK5 activity), and/or exposed to FSS (12 dyn/cm(2)). Cell proliferation was evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The protein expression levels of Gαq, P-ERK5, ERK5, Cyclin B1, and CDK1 were analyzed by Western blot. Physiological FSS exposure for 60 min remarkably promoted MC3T3-E1 cell proliferation, however, this effect was suppressed by siRNA-mediated Gαq knockdown or inhibition of ERK5 activity by XMD8-92 treatment, suggesting that Gαq and ERK5 might modulate FSS-increased osteoblast proliferation. Furthermore, ERK5 phosphorylation was dramatically inhibited by Gαq siRNA. In addition, our study further revealed that FSS treatment of MC3T3-E1 cells for 60 min markedly upregulated the protein expression levels of Cyclin B1 and CDK1, and this increased expression was predominantly blocked by Gαq siRNA or XMD8-92 treatment. We propose that FSS acts on the Gαq-ERK5 signaling pathway to upregulate Cyclin B1 and CDK1 expression, thereby resulting in MC3T3-E1 cell proliferation. Thus, the Gαq-ERK5 signaling pathway may provide useful information regarding the treatment of bone metabolic disease.
Collapse
Affiliation(s)
- Zhang Bo
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Geng Bin
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Wang Jing
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Wang Cuifang
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - An Liping
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Ma Jinglin
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Jiang Jin
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Tan Xiaoyi
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Chen Cong
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Ding Ning
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| | - Xia Yayi
- a Department of Orthopedics , The Second Hospital of Lanzhou University , Lanzhou , China.,b Key Laboratory of Orthopedics of Gansu Province, The Second Hospital of Lanzhou University , Lanzhou , China
| |
Collapse
|
12
|
Simultaneous Increases in Proliferation and Apoptosis of Vascular Smooth Muscle Cells Accelerate Diabetic Mouse Venous Atherosclerosis. PLoS One 2015; 10:e0141375. [PMID: 26488175 PMCID: PMC4619075 DOI: 10.1371/journal.pone.0141375] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 10/06/2015] [Indexed: 12/20/2022] Open
Abstract
Aims This study was designed to demonstrate simultaneous increases in proliferation and apoptosis of vascular smooth muscle cells (VSMCs) leading to accelerated vein graft remodeling and to explore the underlying mechanisms. Methods Vein grafts were performed in non-diabetic and diabetic mice. The cultured quiescent VSMCs were subjected to mechanical stretch stress (SS) and/or advanced glycosylation end products (AGEs). Harvested vein grafts and treated VSMCs were used to detect cell proliferation, apoptosis, mitogen-activated protein kinases (MAPKs) activation and SM-α-actin expression. Results Significantly thicker vessel walls and greater increases in proliferation and apoptosis were observed in diabetic vein grafts than those in non-diabetic. Both SS and AGEs were found to induce different activation of three members of MAPKs and simultaneous increases in proliferation and apoptosis of VSMCs, and combined treatment with both had a synergistic effect. VSMCs with strong SM-α-actin expression represented more activated JNKs or p38MAPK, and cell apoptosis, while the cells with weak SM-α-actin expression demonstrated preferential activation of ERKs and cell proliferation. In contrast, inhibition of MAPKs signals triggered significant decreases in VSMC proliferation, and apoptosis. Treatment of the cells with RNA interference of receptor of AGEs (RAGE) also resulted in significant decreases in both proliferation and apoptosis. Conclusions Increased pressure-induced SS triggers simultaneous increases in proliferation and apoptosis of VSMCs in the vein grafts leading to vein arterializations, which can be synergistically accelerated by high glucose-induced AGEs resulting in vein graft atherosclerosis. Either SS or AGEs and their combination induce simultaneous increases in proliferation and apoptosis of VSMCs via different activation of three members of MAPKs resulting from different VSMC subtypes classified by SM-α-actin expression levels.
Collapse
|
13
|
Variability in vascular smooth muscle cell stretch-induced responses in 2D culture. Vasc Cell 2015; 7:7. [PMID: 26301087 PMCID: PMC4546126 DOI: 10.1186/s13221-015-0032-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 08/12/2015] [Indexed: 01/27/2023] Open
Abstract
The pulsatile nature of blood flow exposes vascular smooth muscle cells (VSMCs) in the vessel wall to mechanical stress, in the form of circumferential and longitudinal stretch. Cyclic stretch evokes VSMC proliferation, apoptosis, phenotypic switching, migration, alignment, and vascular remodeling. Given that these responses have been observed in many cardiovascular diseases, a defined understanding of their underlying mechanisms may provide critical insight into the pathophysiology of cardiovascular derangements. Cyclic stretch-triggered VSMC responses and their effector mechanisms have been studied in vitro using tension systems that apply either uniaxial or equibiaxial stretch to cells grown on an elastomer-bottomed culture plate and ex vivo by stretching whole vein segments with small weights. This review will focus mainly on VSMC responses to the in vitro application of mechanical stress, outlining the inconsistencies in acquired data, and comparing them to in vivo or ex vivo findings. Major discrepancies in data have been seen in mechanical stress-induced proliferation, apoptosis, and phenotypic switching responses, depending on the stretch conditions. These discrepancies stem from variations in stretch conditions such as degree, axis, duration, and frequency of stretch, wave function, membrane coating, cell type, cell passage number, culture media content, and choice of in vitro model. Further knowledge into the variables that cause these incongruities will allow for improvement of the in vitro application of cyclic stretch.
Collapse
|
14
|
Rodríguez AI, Csányi G, Ranayhossaini DJ, Feck DM, Blose KJ, Assatourian L, Vorp DA, Pagano PJ. MEF2B-Nox1 signaling is critical for stretch-induced phenotypic modulation of vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 2015; 35:430-8. [PMID: 25550204 PMCID: PMC4409426 DOI: 10.1161/atvbaha.114.304936] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Blood vessel hemodynamics have profound influences on function and structure of vascular cells. One of the main mechanical forces influencing vascular smooth muscle cells (VSMC) is cyclic stretch (CS). Increased CS stimulates reactive oxygen species (ROS) production in VSMC, leading to their dedifferentiation, yet the mechanisms involved are poorly understood. This study was designed to test the hypothesis that pathological CS stimulates NADPH oxidase isoform 1 (Nox1)-derived ROS via MEF2B, leading to VSMC dysfunction via a switch from a contractile to a synthetic phenotype. APPROACH AND RESULTS Using a newly developed isoform-specific Nox1 inhibitor and gene silencing technology, we demonstrate that a novel pathway, including MEF2B-Nox1-ROS, is upregulated under pathological stretch conditions, and this pathway promotes a VSMC phenotypic switch from a contractile to a synthetic phenotype. We observed that CS (10% at 1 Hz) mimicking systemic hypertension in humans increased Nox1 mRNA, protein levels, and enzymatic activity in a time-dependent manner, and this upregulation was mediated by MEF2B. Furthermore, we show that stretch-induced Nox1-derived ROS upregulated a specific marker for synthetic phenotype (osteopontin), whereas it downregulated classical markers for contractile phenotype (calponin1 and smoothelin B). In addition, our data demonstrated that stretch-induced Nox1 activation decreases actin fiber density and augments matrix metalloproteinase 9 activity, VSMC migration, and vectorial alignment. CONCLUSIONS These results suggest that CS initiates a signal through MEF2B that potentiates Nox1-mediated ROS production and causes VSMC to switch to a synthetic phenotype. The data also characterize a new Nox1 inhibitor as a potential therapy for treatment of vascular dysfunction in hypertension.
Collapse
MESH Headings
- Animals
- Biomarkers/metabolism
- Calcium-Binding Proteins/metabolism
- Cell Movement
- Cells, Cultured
- Cytoskeletal Proteins/metabolism
- Enzyme Inhibitors/pharmacology
- MEF2 Transcription Factors/genetics
- MEF2 Transcription Factors/metabolism
- Matrix Metalloproteinase 9/metabolism
- Mechanotransduction, Cellular/drug effects
- Microfilament Proteins/metabolism
- Muscle Proteins/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- NADH, NADPH Oxidoreductases/antagonists & inhibitors
- NADH, NADPH Oxidoreductases/genetics
- NADH, NADPH Oxidoreductases/metabolism
- NADPH Oxidase 1
- Osteopontin/metabolism
- Phenotype
- Pressoreceptors/metabolism
- RNA Interference
- RNA, Messenger/metabolism
- Rats
- Reactive Oxygen Species/metabolism
- Time Factors
- Transfection
- Vascular Remodeling/drug effects
- Calponins
Collapse
Affiliation(s)
- Andrés I Rodríguez
- From the Department of Pharmacology and Chemical Biology and Vascular Medicine Institute (A.I.R., G.C., D.J.R, D.M.F., L.A., P.J.P), and Departments of Bioengineering, Surgery, and Cardiothoracic Surgery and Center for Vascular Remodeling and Regeneration (K.J.B., D.A.V), University of Pittsburgh, PA; and Department of Basic Sciences, Faculty of Science, Universidad del Bío-Bío, Chillán, Chile (A.I.R)
| | - Gábor Csányi
- From the Department of Pharmacology and Chemical Biology and Vascular Medicine Institute (A.I.R., G.C., D.J.R, D.M.F., L.A., P.J.P), and Departments of Bioengineering, Surgery, and Cardiothoracic Surgery and Center for Vascular Remodeling and Regeneration (K.J.B., D.A.V), University of Pittsburgh, PA; and Department of Basic Sciences, Faculty of Science, Universidad del Bío-Bío, Chillán, Chile (A.I.R)
| | - Daniel J Ranayhossaini
- From the Department of Pharmacology and Chemical Biology and Vascular Medicine Institute (A.I.R., G.C., D.J.R, D.M.F., L.A., P.J.P), and Departments of Bioengineering, Surgery, and Cardiothoracic Surgery and Center for Vascular Remodeling and Regeneration (K.J.B., D.A.V), University of Pittsburgh, PA; and Department of Basic Sciences, Faculty of Science, Universidad del Bío-Bío, Chillán, Chile (A.I.R)
| | - Douglas M Feck
- From the Department of Pharmacology and Chemical Biology and Vascular Medicine Institute (A.I.R., G.C., D.J.R, D.M.F., L.A., P.J.P), and Departments of Bioengineering, Surgery, and Cardiothoracic Surgery and Center for Vascular Remodeling and Regeneration (K.J.B., D.A.V), University of Pittsburgh, PA; and Department of Basic Sciences, Faculty of Science, Universidad del Bío-Bío, Chillán, Chile (A.I.R)
| | - Kory J Blose
- From the Department of Pharmacology and Chemical Biology and Vascular Medicine Institute (A.I.R., G.C., D.J.R, D.M.F., L.A., P.J.P), and Departments of Bioengineering, Surgery, and Cardiothoracic Surgery and Center for Vascular Remodeling and Regeneration (K.J.B., D.A.V), University of Pittsburgh, PA; and Department of Basic Sciences, Faculty of Science, Universidad del Bío-Bío, Chillán, Chile (A.I.R)
| | - Lillian Assatourian
- From the Department of Pharmacology and Chemical Biology and Vascular Medicine Institute (A.I.R., G.C., D.J.R, D.M.F., L.A., P.J.P), and Departments of Bioengineering, Surgery, and Cardiothoracic Surgery and Center for Vascular Remodeling and Regeneration (K.J.B., D.A.V), University of Pittsburgh, PA; and Department of Basic Sciences, Faculty of Science, Universidad del Bío-Bío, Chillán, Chile (A.I.R)
| | - David A Vorp
- From the Department of Pharmacology and Chemical Biology and Vascular Medicine Institute (A.I.R., G.C., D.J.R, D.M.F., L.A., P.J.P), and Departments of Bioengineering, Surgery, and Cardiothoracic Surgery and Center for Vascular Remodeling and Regeneration (K.J.B., D.A.V), University of Pittsburgh, PA; and Department of Basic Sciences, Faculty of Science, Universidad del Bío-Bío, Chillán, Chile (A.I.R)
| | - Patrick J Pagano
- From the Department of Pharmacology and Chemical Biology and Vascular Medicine Institute (A.I.R., G.C., D.J.R, D.M.F., L.A., P.J.P), and Departments of Bioengineering, Surgery, and Cardiothoracic Surgery and Center for Vascular Remodeling and Regeneration (K.J.B., D.A.V), University of Pittsburgh, PA; and Department of Basic Sciences, Faculty of Science, Universidad del Bío-Bío, Chillán, Chile (A.I.R).
| |
Collapse
|
15
|
He W, Li Y, Chen X, Lu L, Tang B, Wang Z, Pan Y, Cai S, He Y, Ke Z. miR-494 acts as an anti-oncogene in gastric carcinoma by targeting c-myc. J Gastroenterol Hepatol 2014; 29:1427-1434. [PMID: 24612089 DOI: 10.1111/jgh.12558] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/20/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND We recently showed that miR-494 was downregulated in gastric carcinoma (GC). The objectives of this study were to determine the role of miR-494 in GC malignancy and to identify its target genes. METHODS Real-time polymerase chain reaction was employed to quantify the expression level of miR-494 and c-myc in gastric cancer tissues. Bioinformatics was used to predict the downstream target genes of miR-494, which were confirmed by luciferase and RNA immunoprecipitation assays. Cell functional analyses and a xenograft mouse model were used to evaluate the role of miR-494 in malignancy. RESULTS miR-494 was downregulated in human GC tissues and in GC cells and was negatively correlated with c-myc expression. High level of c-myc or low level of miR-494 correlated with poor prognosis. The miR-494-binding site in the c-myc 3' untranslated region was predicted using TargetScan and was confirmed by the luciferase assay. Additionally, c-myc and miR-494 were enriched in coimmunoprecipitates with tagged Argonaute2 proteins in cells overexpressing miR-494. Furthermore, a miR-494 mimic significantly downregulated endogenous c-myc expression, which may contribute to the delayed G1/S transition, decreased synthesis phase bromodeoxyuridine incorporation, and impaired cell growth and colony formation; on the other hand, treatment with a miR-494 inhibitor displayed the opposite effects. Reduced tumor burden and decreased cell proliferation were observed following the delivery of miR-494 into xenograft mice. CONCLUSION miR-494 is downregulated in human GC and acts as an anti-oncogene by targeting c-myc. miR-494 plays a role in the pathogenesis of gastric cancer in a recessive fashion.
Collapse
Affiliation(s)
- Weiling He
- Department of Gastrointestinal and Pancreatic Surgery, Centre of Gastric Cancer, The First Affiliated Hospital, Guangzhou, Guangdong, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|