1
|
Niu K, Zhang C, Yang M, Maguire EM, Shi Z, Sun S, Wu J, Liu C, An W, Wang X, Gao S, Ge S, Xiao Q. Small nucleolar RNA host gene 18 controls vascular smooth muscle cell contractile phenotype and neointimal hyperplasia. Cardiovasc Res 2024; 120:796-810. [PMID: 38498586 PMCID: PMC11135647 DOI: 10.1093/cvr/cvae055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 12/27/2023] [Indexed: 03/20/2024] Open
Abstract
AIMS Long non-coding RNA (LncRNA) small nucleolar RNA host gene 18 (SNHG18) has been widely implicated in cancers. However, little is known about its functional involvement in vascular diseases. Herein, we attempted to explore a role for SNHG18 in modulating vascular smooth muscle cell (VSMC) contractile phenotype and injury-induced neointima formation. METHODS AND RESULTS Analysis of single-cell RNA sequencing and transcriptomic datasets showed decreased levels of SNHG18 in injured and atherosclerotic murine and human arteries, which is positively associated with VSMC contractile genes. SNHG18 was upregulated in VSMCs by TGFβ1 through transcription factors Sp1 and SMAD3. SNHG18 gene gain/loss-of-function studies revealed that VSMC contractile phenotype was positively regulated by SNHG18. Mechanistic studies showed that SNHG18 promotes a contractile VSMC phenotype by up-regulating miR-22-3p. SNHG18 up-regulates miR-22 biogenesis and miR-22-3p production by competitive binding with the A-to-I RNA editing enzyme, adenosine deaminase acting on RNA-2 (ADAR2). Surprisingly, we observed that ADAR2 inhibited miR-22 biogenesis not through increasing A-to-I editing within primary miR-22, but by interfering with the binding of microprocessor complex subunit DGCR8 to primary miR-22. Importantly, perivascular SNHG18 overexpression in the injured vessels dramatically up-regulated the expression levels of miR-22-3p and VSMC contractile genes, and prevented injury-induced neointimal hyperplasia. Such modulatory effects were reverted by miR-22-3p inhibition in the injured arteries. Finally, we observed a similar regulator role for SNHG18 in human VSMCs and a decreased expression level of both SNHG18 and miR-22-3p in diseased human arteries; and we found that the expression level of SNHG18 was positively associated with that of miR-22-3p in both healthy and diseased human arteries. CONCLUSION We demonstrate that SNHG18 is a novel regulator in governing VSMC contractile phenotype and preventing injury-induced neointimal hyperplasia. Our findings have important implications for therapeutic targeting snhg18/miR-22-3p signalling in vascular diseases.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Carotid Artery Injuries/pathology
- Carotid Artery Injuries/genetics
- Carotid Artery Injuries/metabolism
- Cells, Cultured
- Disease Models, Animal
- Gene Expression Regulation
- Hyperplasia
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- MicroRNAs/metabolism
- MicroRNAs/genetics
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima
- Phenotype
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA-Binding Proteins/metabolism
- RNA-Binding Proteins/genetics
- Signal Transduction
Collapse
Affiliation(s)
- Kaiyuan Niu
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
- Department of Otorhinolaryngology, Third Affiliated Hospital of Anhui Medical University, No. 390, Huaihe Road, LuYang District, Hefei, Anhui, 230061, PR China
| | - Chengxin Zhang
- Department of Cardiovascular Surgery, First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, Anhui, 230022, PR China
| | - Mei Yang
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Eithne Margaret Maguire
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Zhenning Shi
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Shasha Sun
- Department of Cardiology, Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jianping Wu
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Chenxin Liu
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Weiwei An
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
| | - Xinxin Wang
- Department of Cardiovascular Surgery, First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, Anhui, 230022, PR China
| | - Shan Gao
- Department of Pharmacology, Basic Medical College, Anhui Medical University, No. 81, Meishan Road, Shushan District, Hefei, Anhui, 230032, PR China
| | - Shenglin Ge
- Department of Cardiovascular Surgery, First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, Anhui, 230022, PR China
| | - Qingzhong Xiao
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, John Vane Science Centre, Charterhouse Square, London, EC1M 6BQ, UK
- Department of Cardiovascular Surgery, First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, Anhui, 230022, PR China
- Department of Pharmacology, Basic Medical College, Anhui Medical University, No. 81, Meishan Road, Shushan District, Hefei, Anhui, 230032, PR China
| |
Collapse
|
2
|
Sivaraman S, Ravishankar P, Rao RR. Differentiation and Engineering of Human Stem Cells for Smooth Muscle Generation. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:1-9. [PMID: 35491587 DOI: 10.1089/ten.teb.2022.0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Cardiovascular diseases are responsible for 31% of global deaths and are considered the main cause of death and disability worldwide. Stem cells from various sources have become attractive options for a range of cell-based therapies for smooth muscle tissue regeneration. However, for efficient myogenic differentiation, the stem cell characteristics, cell culture conditions, and their respective microenvironments need to be carefully assessed. This review covers the various approaches involved in the regeneration of vascular smooth muscles by conditioning human stem cells. This article delves into the different sources of stem cells used in the generation of myogenic tissues, the role of soluble growth factors, use of scaffolding techniques, biomolecular cues, relevance of mechanical stimulation, and key transcription factors involved, aimed at inducing myogenic differentiation. Impact statement The review article's main goal is to discuss the recent advances in the field of smooth muscle tissue regeneration. We look at various cell sources, growth factors, scaffolds, mechanical stimuli, and factors involved in smooth muscle formation. These stem cell-based approaches for vascular muscle formation will provide various options for cell-based therapies with long-term beneficial effects on patients.
Collapse
Affiliation(s)
- Srikanth Sivaraman
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| | - Prashanth Ravishankar
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| | - Raj R Rao
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| |
Collapse
|
3
|
Dong CX, Malecki C, Robertson E, Hambly B, Jeremy R. Molecular Mechanisms in Genetic Aortopathy-Signaling Pathways and Potential Interventions. Int J Mol Sci 2023; 24:ijms24021795. [PMID: 36675309 PMCID: PMC9865322 DOI: 10.3390/ijms24021795] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Thoracic aortic disease affects people of all ages and the majority of those aged <60 years have an underlying genetic cause. There is presently no effective medical therapy for thoracic aneurysm and surgery remains the principal intervention. Unlike abdominal aortic aneurysm, for which the inflammatory/atherosclerotic pathogenesis is well established, the mechanism of thoracic aneurysm is less understood. This paper examines the key cell signaling systems responsible for the growth and development of the aorta, homeostasis of endothelial and vascular smooth muscle cells and interactions between pathways. The evidence supporting a role for individual signaling pathways in pathogenesis of thoracic aortic aneurysm is examined and potential novel therapeutic approaches are reviewed. Several key signaling pathways, notably TGF-β, WNT, NOTCH, PI3K/AKT and ANGII contribute to growth, proliferation, cell phenotype and survival for both vascular smooth muscle and endothelial cells. There is crosstalk between pathways, and between vascular smooth muscle and endothelial cells, with both synergistic and antagonistic interactions. A common feature of the activation of each is response to injury or abnormal cell stress. Considerable experimental evidence supports a contribution of each of these pathways to aneurysm formation. Although human information is less, there is sufficient data to implicate each pathway in the pathogenesis of human thoracic aneurysm. As some pathways i.e., WNT and NOTCH, play key roles in tissue growth and organogenesis in early life, it is possible that dysregulation of these pathways results in an abnormal aortic architecture even in infancy, thereby setting the stage for aneurysm development in later life. Given the fine tuning of these signaling systems, functional polymorphisms in key signaling elements may set up a future risk of thoracic aneurysm. Multiple novel therapeutic agents have been developed, targeting cell signaling pathways, predominantly in cancer medicine. Future investigations addressing cell specific targeting, reduced toxicity and also less intense treatment effects may hold promise for effective new medical treatments of thoracic aortic aneurysm.
Collapse
Affiliation(s)
- Charlotte Xue Dong
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Cassandra Malecki
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
| | - Elizabeth Robertson
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Brett Hambly
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Richmond Jeremy
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
- Correspondence:
| |
Collapse
|
4
|
Human Induced Pluripotent Stem Cell-Derived Vascular Cells: Recent Progress and Future Directions. J Cardiovasc Dev Dis 2021; 8:jcdd8110148. [PMID: 34821701 PMCID: PMC8622843 DOI: 10.3390/jcdd8110148] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) hold great promise for cardiovascular regeneration following ischemic injury. Considerable effort has been made toward the development and optimization of methods to differentiate hiPSCs into vascular cells, such as endothelial and smooth muscle cells (ECs and SMCs). In particular, hiPSC-derived ECs have shown robust potential for promoting neovascularization in animal models of cardiovascular diseases, potentially achieving significant and sustained therapeutic benefits. However, the use of hiPSC-derived SMCs that possess high therapeutic relevance is a relatively new area of investigation, still in the earlier investigational stages. In this review, we first discuss different methodologies to derive vascular cells from hiPSCs with a particular emphasis on the role of key developmental signals. Furthermore, we propose a standardized framework for assessing and defining the EC and SMC identity that might be suitable for inducing tissue repair and regeneration. We then highlight the regenerative effects of hiPSC-derived vascular cells on animal models of myocardial infarction and hindlimb ischemia. Finally, we address several obstacles that need to be overcome to fully implement the use of hiPSC-derived vascular cells for clinical application.
Collapse
|
5
|
Wang N, Lu L, Cao QF, Qian S, Ding J, Wang C, Duan H, Shen H, Qi J. Partial inhibition of activin receptor-like kinase 4 alleviates bladder fibrosis caused by bladder outlet obstruction. Exp Cell Res 2021; 406:112724. [PMID: 34237300 DOI: 10.1016/j.yexcr.2021.112724] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/22/2021] [Accepted: 06/29/2021] [Indexed: 12/17/2022]
Abstract
The bladder undergoes profound structural alterations after bladder outlet obstruction (BOO), characterized by hypertrophy of the bladder wall and accumulation of extracellular matrix (ECM). Transforming growth factor-β (TGF-β) has been found to promote fibrosis of the bladder induced by partial bladder outlet obstruction (pBOO). Activin receptor-like kinase 4 (ALK4) is a downstream receptor of the TGF-β superfamily. However, the role of the ALK4-Smad2/3 pathway in the pathogenesis of bladder fibrosis caused by pBOO remains unknown. This study focused on learning the role of ALK4 in the process of bladder fibrosis caused by pBOO. The pBOO mice models showed that ALK4 expression was found to upregulate in the wild-type bladder 6 weeks after pBOO compared to control group. Then, mice with heterozygous knockout of the ALK4 gene (ALK4+/-) were generated. Histological analysis and Western blot (WB) results showed significant suppression of collagen expression in the bladders of ALK4+/- mice after pBOO compared with WT mice. WB also showed that ALK4+/- mice demonstrated significant suppression of phosphorylated Smad2/3 (p-Smad2/3) expression in the bladder 6 weeks after pBOO but not of phosphorylated extracellular signal-regulated kinase, c-Jun N-terminal kinase or protein 38 (p-ERK, p-JNK, p-P38) expression. This effect might have occurred through partial inactivation of the Smad2/3 signaling pathway. In vitro, ALK4 overexpression promoted collagen production in cultured BSMCs and activated the Smad2/3 signaling pathway. Taken together, our results demonstrated that ALK4 insufficiency alleviated bladder fibrosis in a mouse model of pBOO partly by suppressing Smad2/3 activity.
Collapse
Affiliation(s)
- Ning Wang
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, China; Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Lu Lu
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, China
| | - Qi Feng Cao
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Subo Qian
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Jie Ding
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Chen Wang
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Huangqi Duan
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Haibo Shen
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| | - Jun Qi
- Department of Urology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| |
Collapse
|
6
|
Wang X, Ge Y, Shi M, Dai H, Liu W, Wang P. Protein kinase N1 promotes proliferation and invasion of liver cancer. Exp Ther Med 2021; 21:651. [PMID: 33968181 PMCID: PMC8097187 DOI: 10.3892/etm.2021.10083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 03/22/2021] [Indexed: 12/24/2022] Open
Abstract
Protein kinase (PK) N1, also called PKC-related protein 1, participates in the proliferation, invasion and metastasis of various malignant tumors. However, the role of PKN1 in liver cancer remains to be elucidated. The present study investigated the expression of PKN1 using immunohistochemistry in surgical specimens from 36 patients and analyzed the correlation with VEGF, microvascular density (MVD), cell proliferation index (Ki67) and clinicopathological parameters. PKN1 was highly expressed in hepatocellular carcinoma (HCC) and was positively correlated with histological grading of HCC, Ki67 expression and MVD. PKN1 expression in moderately and poorly differentiated HCC was significantly higher compared with highly differentiated HCC. Expression of PKN1 was positively correlated with Ki67 and MVD, and Ki67 expression was positively correlated with MVD. The effects of PKN1 on proliferation, invasion and apoptosis of liver cancer cells were detected in vitro. Cell viability, migration and invasion were reduced and the apoptosis rate was significantly improved when PKN1 expression was silenced in liver cancer cells. Thus, PKN1 serves an important role in the development and progression of liver cancer. Inhibition of PKN1 activity may provide a promising therapeutic target for liver cancer.
Collapse
Affiliation(s)
- Xia Wang
- Department of Pathology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Yansong Ge
- Department of Radiology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Mingqi Shi
- Department of Radiology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Hanhan Dai
- Department of Radiology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Wei Liu
- Department of Radiology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Peiyuan Wang
- Department of Radiology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| |
Collapse
|
7
|
Bonetti J, Corti A, Lerouge L, Pompella A, Gaucher C. Phenotypic Modulation of Macrophages and Vascular Smooth Muscle Cells in Atherosclerosis-Nitro-Redox Interconnections. Antioxidants (Basel) 2021; 10:antiox10040516. [PMID: 33810295 PMCID: PMC8066740 DOI: 10.3390/antiox10040516] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
Monocytes/macrophages and vascular smooth muscle cells (vSMCs) are the main cell types implicated in atherosclerosis development, and unlike other mature cell types, both retain a remarkable plasticity. In mature vessels, differentiated vSMCs control the vascular tone and the blood pressure. In response to vascular injury and modifications of the local environment (inflammation, oxidative stress), vSMCs switch from a contractile to a secretory phenotype and also display macrophagic markers expression and a macrophagic behaviour. Endothelial dysfunction promotes adhesion to the endothelium of monocytes, which infiltrate the sub-endothelium and differentiate into macrophages. The latter become polarised into M1 (pro-inflammatory), M2 (anti-inflammatory) or Mox macrophages (oxidative stress phenotype). Both monocyte-derived macrophages and macrophage-like vSMCs are able to internalise and accumulate oxLDL, leading to formation of “foam cells” within atherosclerotic plaques. Variations in the levels of nitric oxide (NO) can affect several of the molecular pathways implicated in the described phenomena. Elucidation of the underlying mechanisms could help to identify novel specific therapeutic targets, but to date much remains to be explored. The present article is an overview of the different factors and signalling pathways implicated in plaque formation and of the effects of NO on the molecular steps of the phenotypic switch of macrophages and vSMCs.
Collapse
Affiliation(s)
- Justine Bonetti
- CITHEFOR, Université de Lorraine, F-54000 Nancy, France; (J.B.); (L.L.); (C.G.)
| | - Alessandro Corti
- Department of Translational Research NTMS, University of Pisa Medical School, 56126 Pisa, Italy;
| | - Lucie Lerouge
- CITHEFOR, Université de Lorraine, F-54000 Nancy, France; (J.B.); (L.L.); (C.G.)
| | - Alfonso Pompella
- Department of Translational Research NTMS, University of Pisa Medical School, 56126 Pisa, Italy;
- Correspondence: ; Tel.: +39-050-2218-537
| | - Caroline Gaucher
- CITHEFOR, Université de Lorraine, F-54000 Nancy, France; (J.B.); (L.L.); (C.G.)
| |
Collapse
|
8
|
The structure and function of protein kinase C-related kinases (PRKs). Biochem Soc Trans 2021; 49:217-235. [PMID: 33522581 PMCID: PMC7925014 DOI: 10.1042/bst20200466] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/29/2020] [Accepted: 01/07/2021] [Indexed: 11/17/2022]
Abstract
The protein kinase C-related kinase (PRK) family of serine/threonine kinases, PRK1, PRK2 and PRK3, are effectors for the Rho family small G proteins. An array of studies have linked these kinases to multiple signalling pathways and physiological roles, but while PRK1 is relatively well-characterized, the entire PRK family remains understudied. Here, we provide a holistic overview of the structure and function of PRKs and describe the molecular events that govern activation and autoregulation of catalytic activity, including phosphorylation, protein interactions and lipid binding. We begin with a structural description of the regulatory and catalytic domains, which facilitates the understanding of their regulation in molecular detail. We then examine their diverse physiological roles in cytoskeletal reorganization, cell adhesion, chromatin remodelling, androgen receptor signalling, cell cycle regulation, the immune response, glucose metabolism and development, highlighting isoform redundancy but also isoform specificity. Finally, we consider the involvement of PRKs in pathologies, including cancer, heart disease and bacterial infections. The abundance of PRK-driven pathologies suggests that these enzymes will be good therapeutic targets and we briefly report some of the progress to date.
Collapse
|
9
|
PKN2 is involved in aggregation and spheroid formation of fibroblasts in suspension culture by regulating cell motility and N-cadherin expression. Biochem Biophys Rep 2021; 25:100895. [PMID: 33437883 PMCID: PMC7787963 DOI: 10.1016/j.bbrep.2020.100895] [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: 11/22/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 01/04/2023] Open
Abstract
The role of Protein Kinase N2 (PKN2, also known as PRK2/PKNγ) in cell aggregate/spheroid formation in suspension culture was investigated using immortalized fibroblasts established from PKN2flox/flox mouse embryos. PKN2flox/flox cells formed cell aggregates in flat bottom low attachment well plates, such as 2% agar and poly-2-hydroxyethymethacrylate coated plates, however, Cre;PKN2flox/flox cells in which PKN2 was depleted by the introduction of Cre-recombinase rarely formed aggregates. Time-lapse analysis revealed that the velocity of Cre;PKN2flox/flox cell motility was significantly lower than that of PKN2flox/flox in a low attachment flat-bottom plate, which likely resulted in a lower cell-cell contact frequency among Cre;PKN2flox/flox cells. Conversely, Cre;PKN2flox/flox cells could form initial cell aggregates in U-bottom low attachment well plates, however, the succeeding compaction process was delayed in Cre;PKN2flox/flox cells with decreased roundness, although PKN2flox/flox cells underwent compaction in a round shape spheroid within 24 h. Immunoblot analysis revealed that the preparation of the cell suspension from adherent conditions using trypsin/EDTA treatment significantly decreased the expression of N-cadherin in both PKN2flox/flox and Cre;PKN2flox/flox cells. The N-cadherin expression level recovered time-dependently; however, the recovery of N-cadherin was significantly delayed in Cre;PKN2flox/flox cells compared to PKN2flox/flox cells. Reverse transcription quantitative PCR revealed that N-cadherin mRNA in Cre;PKN2flox/flox cells was significantly lower than that of PKN2flox/flox cells. These results suggest that PKN2 controls the velocity of cell motility and the transcription of N-cadherin in fibroblasts, leading to cell aggregation and compaction for spheroid formation in suspension culture. PKN2 is involved in initial fibroblast aggregation by regulating cell motility. PKN2 is involved in compaction of fibroblasts in suspension. N-cadherin protein level seems to be a key element for compaction of fibroblasts. PKN2 controls transcription of N-cadherin mRNA in fibroblasts.
Collapse
|
10
|
Garrison CM, Singh-Varma A, Pastino AK, Steele JAM, Kohn J, Murthy NS, Schwarzbauer JE. A multilayered scaffold for regeneration of smooth muscle and connective tissue layers. J Biomed Mater Res A 2020; 109:733-744. [PMID: 32654327 DOI: 10.1002/jbm.a.37058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/18/2020] [Accepted: 06/23/2020] [Indexed: 01/26/2023]
Abstract
Tissue regeneration often requires recruitment of different cell types and rebuilding of two or more tissue layers to restore function. Here, we describe the creation of a novel multilayered scaffold with distinct fiber organizations-aligned to unaligned and dense to porous-to template common architectures found in adjacent tissue layers. Electrospun scaffolds were fabricated using a biodegradable, tyrosine-derived terpolymer, yielding densely-packed, aligned fibers that transition into randomly-oriented fibers of increasing diameter and porosity. We demonstrate that differently-oriented scaffold fibers direct cell and extracellular matrix (ECM) organization, and that scaffold fibers and ECM protein networks are maintained after decellularization. Smooth muscle and connective tissue layers are frequently adjacent in vivo; we show that within a single scaffold, the architecture supports alignment of contractile smooth muscle cells and deposition by fibroblasts of a meshwork of ECM fibrils. We rolled a flat scaffold into a tubular construct and, after culture, showed cell viability, orientation, and tissue-specific protein expression in the tube were similar to the flat-sheet scaffold. This scaffold design not only has translational potential for reparation of flat and tubular tissue layers but can also be customized for alternative applications by introducing two or more cell types in different combinations.
Collapse
Affiliation(s)
- Carly M Garrison
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Anya Singh-Varma
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Alexandra K Pastino
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Joseph A M Steele
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Joachim Kohn
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - N Sanjeeva Murthy
- New Jersey Center for Biomaterials, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Jean E Schwarzbauer
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| |
Collapse
|
11
|
PKN1 kinase-negative knock-in mice develop splenomegaly and leukopenia at advanced age without obvious autoimmune-like phenotypes. Sci Rep 2019; 9:13977. [PMID: 31562379 PMCID: PMC6764976 DOI: 10.1038/s41598-019-50419-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 08/30/2019] [Indexed: 01/08/2023] Open
Abstract
Protein kinase N1 (PKN1) knockout (KO) mice spontaneously form germinal centers (GCs) and develop an autoimmune-like disease with age. Here, we investigated the function of PKN1 kinase activity in vivo using aged mice deficient in kinase activity resulting from the introduction of a point mutation (T778A) in the activation loop of the enzyme. PKN1[T778A] mice reached adulthood without external abnormalities; however, the average spleen size and weight of aged PKN1[T778A] mice increased significantly compared to aged wild type (WT) mice. Histologic examination and Southern blot analyses of spleens showed extramedullary hematopoiesis and/or lymphomagenesis in some cases, although without significantly different incidences between PKN1[T778A] and WT mice. Additionally, flow cytometry revealed increased numbers in B220+, CD3+, Gr1+ and CD193+ leukocytes in the spleen of aged PKN1[T778A] mice, whereas the number of lymphocytes, neutrophils, eosinophils, and monocytes was reduced in the peripheral blood, suggesting an advanced impairment of leukocyte trafficking with age. Moreover, aged PKN1[T778A] mice showed no obvious GC formation nor autoimmune-like phenotypes, such as glomerulonephritis or increased anti-dsDNA antibody titer, in peripheral blood. Our results showing phenotypic differences between aged Pkn1-KO and PKN1[T778A] mice may provide insight into the importance of PKN1-specific kinase-independent functions in vivo.
Collapse
|
12
|
Zeng M, Luo Y, Xu C, Li R, Chen N, Deng X, Fang D, Wang L, Wu J, Luo M. Platelet-endothelial cell interactions modulate smooth muscle cell phenotype in an in vitro model of type 2 diabetes mellitus. Am J Physiol Cell Physiol 2018; 316:C186-C197. [PMID: 30517030 DOI: 10.1152/ajpcell.00428.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Platelet (PLT)-endothelial cell (EC) interaction appears to contribute to phenotypic transition of vascular smooth muscle cells (VSMCs), which play an important role in the physiological and pathological process of vascular complications in type 2 diabetes mellitus (DM2). However, the precise mechanisms by which interactions between PLTs and ECs affect VSMC phenotype have largely remained unclear. We determined the effect of diabetic PLT-EC interaction to influence VSMC migration, proliferation, and phenotypic transformation in triple-cell coculture models using the quantitative real-time PCR, Western blot, fluorescence microscopy, wound scratch assays, CCK-8 assays, and gelatin zymography assays. Our results revealed DM2 PLT-EC interaction to be associated with a significant downregulation of VSMC-specific contractile phenotypic genes and proteins, including SM22α, smooth muscle actin, Smoothelin-B, and smooth muscle-myosin heavy chain. Inversely, VSMC-specific proliferative phenotype gene and protein levels, including cyclin D1 and 2, nonmuscle myosin heavy chain B, and PCNA were in upregulation. Furthermore, the DM2-originated PLT-EC interaction promoted the expression level of transforming growth factor-β1, and the PI3K/Akt and matrix metalloproteinase 9 signaling pathway was activated subsequently. Finally, these reactions contributed to a synthetic phenotype of VSMCs, including the proliferation, migration, and gelatinolytic activities. These findings suggest that PLT-EC interaction modulates the phenotypic transition of VSMCs between a contractile and proliferative/synthetic phenotype under diabetic conditions, conceivably providing important implications regarding the mechanisms controlling the VSMC phenotypic transition and the development of cardiovascular complications.
Collapse
Affiliation(s)
- Min Zeng
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China.,Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yulin Luo
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China.,Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,GCP Center, Affiliated Hospital (Traditional Chinese Medicine) of Southwest Medical University, Luzhou, China
| | - Chunrong Xu
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China.,Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Rong Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China.,Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Ni Chen
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China.,Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xin Deng
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China.,Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Dan Fang
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China.,Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Liqun Wang
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China.,Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jianbo Wu
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China.,Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Dalton Cardiovascular Research Center, University of Missouri-Columbia , Columbia, Missouri
| | - Mao Luo
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, China.,Drug Discovery Research Center, Southwest Medical University, Luzhou, China.,Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
13
|
He J, Zhong X, Zhao L, Gan H. JAK2/STAT3/BMP-2 axis and NF-κB pathway are involved in erythropoietin-induced calcification in rat vascular smooth muscle cells. Clin Exp Nephrol 2018; 23:501-512. [PMID: 30406500 DOI: 10.1007/s10157-018-1666-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 10/28/2018] [Indexed: 01/31/2023]
Abstract
BACKGROUND Vascular calcification is common in chronic kidney disease (CKD) patients, while erythropoietin (EPO) is widely used in the treatment of renal anemia in CKD patients, whether there is a link between the two is still not clear. METHODS The primary rat vascular smooth muscle cells (VSMCs) and CKD rats were treated with EPO and the calcium deposition was observed by alizarin red staining, von Kossa staining and calcium quantification. Activation of JAK2/STAT3/BMP-2 axis and NF-κB signaling pathways was investigated by Western blotting. RESULTS EPO-induced calcium deposition in VSMCs and significantly potentiated calcification in CKD rats. Furthermore, EPO activated JAK2/STAT3/BMP-2 axis, NF-κB pathway and the pro-calcification effect of EPO was partially blocked by the STAT3 inhibitor (Cryptotanshinone) or NF-κB inhibitor (BAY 11-7082), respectively, in vitro. CONCLUSION EPO could promote VSMCs calcification in vitro and in vivo and this effect may be achieved through the JAK2/STAT3/BMP-2 axis and NF-κB pathway.
Collapse
Affiliation(s)
- Jin He
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xiaoyi Zhong
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Lin Zhao
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Hua Gan
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
14
|
Gu W, Hong X, Potter C, Qu A, Xu Q. Mesenchymal stem cells and vascular regeneration. Microcirculation 2018; 24. [PMID: 27681821 DOI: 10.1111/micc.12324] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/20/2016] [Indexed: 12/22/2022]
Abstract
In recent years, MSCs have emerged as a promising therapeutic cell type in regenerative medicine. They hold great promise for treating cardiovascular diseases, such as myocardial infarction and limb ischemia. MSCs may be utilized in both cell-based therapy and vascular graft engineering to restore vascular function, thereby providing therapeutic benefits to patients. The efficacy of MSCs lies in their multipotent differentiation ability toward vascular smooth muscle cells, endothelial cells and other cell types, as well as their capacity to secrete various trophic factors, which are potent in promoting angiogenesis, inhibiting apoptosis and modulating immunoreaction. Increasing our understanding of the mechanisms of MSC involvement in vascular regeneration will be beneficial in boosting present therapeutic approaches and developing novel ones to treat cardiovascular diseases. In this review, we aim to summarize current progress in characterizing the in vivo identity of MSCs, to discuss mechanisms involved in cell-based therapy utilizing MSCs, and to explore current and future strategies for vascular regeneration.
Collapse
Affiliation(s)
- Wenduo Gu
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Xuechong Hong
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Claire Potter
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China
| | - Qingbo Xu
- Cardiovascular Division, King's College London BHF Centre, London, UK
| |
Collapse
|
15
|
Goumans MJ, Ten Dijke P. TGF-β Signaling in Control of Cardiovascular Function. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a022210. [PMID: 28348036 DOI: 10.1101/cshperspect.a022210] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Genetic studies in animals and humans indicate that gene mutations that functionally perturb transforming growth factor β (TGF-β) signaling are linked to specific hereditary vascular syndromes, including Osler-Rendu-Weber disease or hereditary hemorrhagic telangiectasia and Marfan syndrome. Disturbed TGF-β signaling can also cause nonhereditary disorders like atherosclerosis and cardiac fibrosis. Accordingly, cell culture studies using endothelial cells or smooth muscle cells (SMCs), cultured alone or together in two- or three-dimensional cell culture assays, on plastic or embedded in matrix, have shown that TGF-β has a pivotal effect on endothelial and SMC proliferation, differentiation, migration, tube formation, and sprouting. Moreover, TGF-β can stimulate endothelial-to-mesenchymal transition, a process shown to be of key importance in heart valve cushion formation and in various pathological vascular processes. Here, we discuss the roles of TGF-β in vasculogenesis, angiogenesis, and lymphangiogenesis and the deregulation of TGF-β signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- Marie-José Goumans
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Peter Ten Dijke
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
16
|
Progression and Characterization of the Accelerated Atherosclerosis in Iliac Artery of New Zealand White Rabbits: Effect of Simvastatin. J Cardiovasc Pharmacol 2018; 69:314-325. [PMID: 28207427 DOI: 10.1097/fjc.0000000000000477] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Although atherosclerosis is described in New Zealand White rabbit's iliac artery, yet details of time-dependent atherosclerosis progression are not well known. Further, a well characterized accelerated model of atherosclerosis is also required for the screening of candidate drugs to target specific steps of atherosclerosis development. The present study extensively characterizes the time-dependent plaque composition and functional responses of the atherosclerosis in rabbit iliac artery and its modification by simvastatin. METHODS Atherosclerosis was induced with a combination of balloon injury and atherogenic diet (AD) (1% cholesterol, 6% peanut oil) in rabbit's iliac artery. Atherosclerosis progression was evaluated on days 8, 10, 15, 21, 35, and 56 after AD feeding. The plaque characterization was done using histology, real-time reverse transcription-polymerase chain reaction, and vasoreactivity experiments. The standard anti-hyperlipidemic drug, simvastatin (5 mg·kg·d), was used to investigate its effect on atherosclerotic changes. RESULTS Plasma lipids were elevated in a progressive manner after AD feeding from days 8 to 56. Similarly, arterial lipids, Monocyte Chemoattractant Protein-1 (MCP-1) level along with infiltration of macrophages in the lesion area were also increased from day 15 onward. This resulted in a significant increase in the plaque area and intimal-medial thickness ratio in contrast to normal animals. Inflammatory milieu was observed with a significant increase in expression of pro-inflammatory regulators like MCP-1, Tumor Necrosis Factor-α (TNF-α) and Vascular Cell Adhesion Molecule-1 (VCAM-1), whereas anti-inflammatory cytokine interleukin 10 decreased as disease progressed. Endothelial dysfunction was also observed, specifically Acetylcholine (ACh)-induced vasorelaxation was reduced from day 8 onward, whereas the phenylephrine-induced vasoconstriction response was progressively reduced from day 15 in the iliac artery. Ground substances including proteoglycans, α-actin, and collagen content along with metalloproteinase-9 and Tissue inhibitor of metalloproteinases-1 (TIMP-1) inhibitors were significantly augmented at later time points, day 21 onward. Simvastatin treatment for 35 days, at a dose having no significant effect on plasma lipid levels, significantly reduced atherosclerotic progression as evident by reduced macrophage content, inflammatory burden, and extracellular matrix component like proteoglycans and metalloproteinase-9. CONCLUSIONS The authors observed that AD feeding with balloon injury in the rabbit iliac artery accelerated the progression of atherosclerosis and exhibited predominant features of type III human lesion within 8 weeks (56 days). Simvastatin treatment for 35 days exhibited anti-atherosclerotic efficacy without significantly lowering the circulating lipids. The current study thus provides an insight into the time-dependent atherosclerotic progression in rabbit iliac artery and highlights its utility for anti-atherosclerotic evaluation of the candidate drugs.
Collapse
|
17
|
Miranda MZ, Bialik JF, Speight P, Dan Q, Yeung T, Szászi K, Pedersen SF, Kapus A. TGF-β1 regulates the expression and transcriptional activity of TAZ protein via a Smad3-independent, myocardin-related transcription factor-mediated mechanism. J Biol Chem 2017; 292:14902-14920. [PMID: 28739802 DOI: 10.1074/jbc.m117.780502] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 06/30/2017] [Indexed: 12/20/2022] Open
Abstract
Hippo pathway transcriptional coactivators TAZ and YAP and the TGF-β1 (TGFβ) effector Smad3 regulate a common set of genes, can physically interact, and exhibit multilevel cross-talk regulating cell fate-determining and fibrogenic pathways. However, a key aspect of this cross-talk, TGFβ-mediated regulation of TAZ or YAP expression, remains uncharacterized. Here, we show that TGFβ induces robust TAZ but not YAP protein expression in both mesenchymal and epithelial cells. TAZ levels, and to a lesser extent YAP levels, also increased during experimental kidney fibrosis. Pharmacological or genetic inhibition of Smad3 did not prevent the TGFβ-induced TAZ up-regulation, indicating that this canonical pathway is dispensable. In contrast, inhibition of p38 MAPK, its downstream effector MK2 (e.g. by the clinically approved antifibrotic pirferidone), or Akt suppressed the TGFβ-induced TAZ expression. Moreover, TGFβ elevated TAZ mRNA in a p38-dependent manner. Myocardin-related transcription factor (MRTF) was a central mediator of this effect, as MRTF silencing/inhibition abolished the TGFβ-induced TAZ expression. MRTF overexpression drove the TAZ promoter in a CC(A/T-rich)6GG (CArG) box-dependent manner and induced TAZ protein expression. TGFβ did not act by promoting nuclear MRTF translocation; instead, it triggered p38- and MK2-mediated, Nox4-promoted MRTF phosphorylation and activation. Functionally, higher TAZ levels increased TAZ/TEAD-dependent transcription and primed cells for enhanced TAZ activity upon a second stimulus (i.e. sphingosine 1-phosphate) that induced nuclear TAZ translocation. In conclusion, our results uncover an important aspect of the cross-talk between TGFβ and Hippo signaling, showing that TGFβ induces TAZ via a Smad3-independent, p38- and MRTF-mediated and yet MRTF translocation-independent mechanism.
Collapse
Affiliation(s)
- Maria Zena Miranda
- From the Keenan Research Centre for Biomedical Science of the St. Michael's Hospital.,Biochemistry, University of Toronto, Toronto, Ontario M5B 1T8N, Canada and
| | - Janne Folke Bialik
- From the Keenan Research Centre for Biomedical Science of the St. Michael's Hospital.,the Department of Cell and Developmental Biology, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Pam Speight
- From the Keenan Research Centre for Biomedical Science of the St. Michael's Hospital
| | - Qinghong Dan
- From the Keenan Research Centre for Biomedical Science of the St. Michael's Hospital
| | - Tony Yeung
- From the Keenan Research Centre for Biomedical Science of the St. Michael's Hospital
| | - Katalin Szászi
- From the Keenan Research Centre for Biomedical Science of the St. Michael's Hospital.,Departments of Surgery and
| | - Stine F Pedersen
- the Department of Cell and Developmental Biology, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - András Kapus
- From the Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, .,Biochemistry, University of Toronto, Toronto, Ontario M5B 1T8N, Canada and.,Departments of Surgery and
| |
Collapse
|
18
|
PPARβ/δ, a Novel Regulator for Vascular Smooth Muscle Cells Phenotypic Modulation and Vascular Remodeling after Subarachnoid Hemorrhage in Rats. Sci Rep 2017; 7:45234. [PMID: 28327554 PMCID: PMC5361085 DOI: 10.1038/srep45234] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 02/20/2017] [Indexed: 12/20/2022] Open
Abstract
Cerebral vascular smooth muscle cell (VSMC) phenotypic switch is involved in the pathophysiology of vascular injury after aneurysmal subarachnoid hemorrhage (aSAH), whereas the molecular mechanism underlying it remains largely speculative. Peroxisome proliferator-activated receptor β/δ (PPARβ/δ) has been implicated to modulate the vascular cells proliferation and vascular homeostasis. In the present study, we investigated the potential role of PPARβ/δ in VSMC phenotypic switch following SAH. Activation of PPARβ/δ by GW0742 and adenoviruses PPARβ/δ (Ad-PPARβ/δ) significantly inhibited hemoglobin-induced VSMC phenotypic switch. However, the effects of PPARβ/δ on VSMC phenotypic switch were partly obstacled in the presence of LY294002, a potent inhibitor of Phosphatidyl-Inositol-3 Kinase-AKT (PI3K/AKT). Furthermore, following study demonstrated that PPARβ/δ-induced PI3K/AKT activation can also contribute to Serum Response Factor (SRF) nucleus localization and Myocardin expression, which was highly associated with VSMC phenotypic switch. Finally, we found that Ad-PPARβ/δ positively modulated vascular remodeling in SAH rats, i.e. the diameter of basilar artery and the thickness of vessel wall. In addition, overexpression of PPARβ/δ by adenoviruses significantly improved neurological outcome. Taken together, this study identified PPARβ/δ as a useful regulator for VSMC phenotypic switch and vascular remodeling following SAH, providing novel insights into the therapeutic strategies of delayed cerebral ischemia.
Collapse
|
19
|
Wang F, Zhan R, Chen L, Dai X, Wang W, Guo R, Li X, Li Z, Wang L, Huang S, Shen J, Li S, Cao C. RhoA promotes epidermal stem cell proliferation via PKN1-cyclin D1 signaling. PLoS One 2017; 12:e0172613. [PMID: 28222172 PMCID: PMC5319766 DOI: 10.1371/journal.pone.0172613] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 02/06/2017] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVE Epidermal stem cells (ESCs) play a critical role in wound healing, but the mechanism underlying ESC proliferation is not well defined. Here, we explore the effects of RhoA on ESC proliferation and the possible underlying mechanism. METHODS Human ESCs were enriched by rapid adhesion to collagen IV. RhoA(+/+)(G14V), RhoA(-/-)(T19N) and pGFP control plasmids were transfected into human ESCs. The effect of RhoA on cell proliferation was detected by cell proliferation and DNA synthesis assays. Induction of PKN1 activity by RhoA was determined by immunoblot analysis, and the effects of PKN1 on RhoA in terms of inducing cell proliferation and cyclin D1 expression were detected using specific siRNA targeting PKN1. The effects of U-46619 (a RhoA agonist) and C3 transferase (a RhoA antagonist) on ESC proliferation were observed in vivo. RESULTS RhoA had a positive effect on ESC proliferation, and PKN1 activity was up-regulated by the active RhoA mutant (G14V) and suppressed by RhoA T19N. Moreover, the ability of RhoA to promote ESC proliferation and DNA synthesis was interrupted by PKN1 siRNA. Additionally, cyclin D1 protein and mRNA expression levels were up-regulated by RhoA G14V, and these effects were inhibited by siRNA-mediated knock-down of PKN1. RhoA also promoted ESC proliferation via PKN in vivo. CONCLUSION This study shows that the effect of RhoA on ESC proliferation is mediated by activation of the PKN1-cyclin D1 pathway in vitro, suggesting that RhoA may serve as a new therapeutic target for wound healing.
Collapse
Affiliation(s)
- Fan Wang
- Department of Plastic and Reconstructive Surgery, Southwestern Hospital, Third Military Medical University, Chongqing, China
| | - Rixing Zhan
- School of Nursing, Third Military Medical University, Chongqing, China
| | - Liang Chen
- Department of Plastic and Reconstructive Surgery, Southwestern Hospital, Third Military Medical University, Chongqing, China
| | - Xia Dai
- Department of Plastic and Reconstructive Surgery, Southwestern Hospital, Third Military Medical University, Chongqing, China
| | - Wenping Wang
- Department of Plastic and Reconstructive Surgery, Southwestern Hospital, Third Military Medical University, Chongqing, China
| | - Rui Guo
- Department of Plastic and Reconstructive Surgery, Southwestern Hospital, Third Military Medical University, Chongqing, China
| | - Xiaoge Li
- Department of Plastic and Reconstructive Surgery, Southwestern Hospital, Third Military Medical University, Chongqing, China
| | - Zhe Li
- Department of Plastic and Reconstructive Surgery, Southwestern Hospital, Third Military Medical University, Chongqing, China
| | - Liang Wang
- Department of Plastic and Reconstructive Surgery, Southwestern Hospital, Third Military Medical University, Chongqing, China
| | - Shupeng Huang
- Department of Plastic and Reconstructive Surgery, Southwestern Hospital, Third Military Medical University, Chongqing, China
| | - Jie Shen
- Department of Plastic and Reconstructive Surgery, Southwestern Hospital, Third Military Medical University, Chongqing, China
| | - Shirong Li
- Department of Plastic and Reconstructive Surgery, Southwestern Hospital, Third Military Medical University, Chongqing, China
- * E-mail: (LS); (CC)
| | - Chuan Cao
- Department of Plastic and Reconstructive Surgery, Southwestern Hospital, Third Military Medical University, Chongqing, China
- * E-mail: (LS); (CC)
| |
Collapse
|
20
|
Xu JG, Zhu SY, Heng BC, Dissanayaka WL, Zhang CF. TGF-β1-induced differentiation of SHED into functional smooth muscle cells. Stem Cell Res Ther 2017; 8:10. [PMID: 28114966 PMCID: PMC5260045 DOI: 10.1186/s13287-016-0459-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 12/02/2016] [Accepted: 12/16/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Adequate vascularization is crucial for supplying nutrition and discharging metabolic waste in freshly transplanted tissue-engineered constructs. Obtaining the appropriate building blocks for vascular tissue engineering (i.e. endothelial and mural cells) is a challenging task for tissue neovascularization. Hence, we investigated whether stem cells from human exfoliated deciduous teeth (SHED) could be induced to differentiate into functional vascular smooth muscle cells (vSMCs). METHODS We utilized two cytokines of the TGF-β family, transforming growth factor beta 1 (TGF-β1) and bone morphogenetic protein 4 (BMP4), to induce SHED differentiation into SMCs. Quantitative real-time polymerase chain reaction (RT-qPCR) was used to assess mRNA expression, and protein expression was analyzed using flow cytometry, western blot and immunostaining. Additionally, to examine whether these SHED-derived SMCs possess the same function as primary SMCs, in vitro Matrigel angiogenesis assay, fibrin gel bead assay, and functional contraction study were used here. RESULTS By analyzing the expression of specific markers of SMCs (α-SMA, SM22α, Calponin, and SM-MHC), we confirmed that TGF-β1, and not BMP4, could induce SHED differentiation into SMCs. The differentiation efficiency was relatively high (α-SMA+ 86.1%, SM22α+ 93.9%, Calponin+ 56.8%, and SM-MHC+ 88.2%) as assessed by flow cytometry. In vitro Matrigel angiogenesis assay showed that the vascular structures generated by SHED-derived SMCs and human umbilical vein endothelial cells (HUVECs) were comparable to primary SMCs and HUVECs in terms of vessel stability. Fibrin gel bead assay showed that SHED-derived SMCs had a stronger capacity for promoting vessel formation compared with primary SMCs. Further analyses of protein expression in fibrin gel showed that cultures containing SHED-derived SMCs exhibited higher expression levels of Fibronectin than the primary SMCs group. Additionally, it was also confirmed that SHED-derived SMCs exhibited functional contractility. When SB-431542, a specific inhibitor of ALK5 was administered, TGF-β1 stimulation could not induce SHED into SMCs, indicating that the differentiation of SHED into SMCs is somehow related to the TGF-β1-ALK5 signaling pathway. CONCLUSIONS SHED could be successfully induced into functional SMCs for vascular tissue engineering, and this course could be regulated through the ALK5 signaling pathway. Hence, SHED appear to be a promising candidate cell type for vascular tissue engineering.
Collapse
Affiliation(s)
- Jian Guang Xu
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
| | - Shao Yue Zhu
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
| | - Boon Chin Heng
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
| | - Waruna Lakmal Dissanayaka
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
- HKU Shenzhen Institute of Research and Innovation, Hong Kong, China
| | - Cheng Fei Zhang
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
- HKU Shenzhen Institute of Research and Innovation, Hong Kong, China
| |
Collapse
|
21
|
Fujishima Y, Maeda N, Matsuda K, Masuda S, Mori T, Fukuda S, Sekimoto R, Yamaoka M, Obata Y, Kita S, Nishizawa H, Funahashi T, Ranscht B, Shimomura I. Adiponectin association with T-cadherin protects against neointima proliferation and atherosclerosis. FASEB J 2017; 31:1571-1583. [PMID: 28062540 DOI: 10.1096/fj.201601064r] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/19/2016] [Indexed: 12/20/2022]
Abstract
Adiponectin, an adipocyte-derived protein abundant in the circulation, is thought to be protective against atherosclerosis. However, it is not fully understood how the association of adiponectin with vascular cells and its antiatherogenic effect are connected. In this study, T-cadherin was essential for accumulation of adiponectin in the neointima and atherosclerotic plaque lesions, and the adiponectin-T-cadherin association protected against vascular injury. In the apolipoprotein E-knockout (ApoE-KO) mice, adiponectin and T-cadherin colocalized on endothelial cells and synthetic smooth muscle cells in the aortic intima. Notably, aortic adiponectin protein disappeared in T-cadherin/ApoE double-knockout (Tcad/ApoE-DKO) mice with significant elevation of blood adiponectin concentration. Furthermore, in Tcad/ApoE-DKO mice, carotid artery ligation resulted in a significant increase of neointimal thickness compared with ApoE-KO mice. Finally, on a high-cholesterol diet, Tcad/ApoE-DKO mice increased atherosclerotic plaque formation, despite a 5-fold increase in plasma adiponectin level compared with that in ApoE-KO mice. In vitro, knockdown of T-cadherin from human aortic smooth muscle cells (HASMCs) with synthetic phenotype significantly reduced adiponectin accumulation on HASMCs and negated the inhibitory effect of adiponectin on proinflammatory change. Collective evidence showed that adiponectin accumulates in the vasculature via T-cadherin, and the adiponectin-T-cadherin association plays a protective role against neointimal and atherosclerotic plaque formations.-Fujishima, Y., Maeda, N., Matsuda, K., Masuda, S., Mori, T., Fukuda, S., Sekimoto, R., Yamaoka, M., Obata, Y., Kita, S., Nishizawa, H., Funahashi, T., Ranscht, B., Shimomura, I. Adiponectin association with T-cadherin protects against neointima proliferation and atherosclerosis.
Collapse
Affiliation(s)
- Yuya Fujishima
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Norikazu Maeda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; .,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Osaka, Japan; and
| | - Keisuke Matsuda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shigeki Masuda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takuya Mori
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shiro Fukuda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Ryohei Sekimoto
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masaya Yamaoka
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshinari Obata
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shunbun Kita
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Osaka, Japan; and
| | - Hitoshi Nishizawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tohru Funahashi
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Osaka, Japan; and
| | - Barbara Ranscht
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
22
|
Song SH, Kim K, Jo EK, Kim YW, Kwon JS, Bae SS, Sung JH, Park SG, Kim JT, Suh W. Fibroblast Growth Factor 12 Is a Novel Regulator of Vascular Smooth Muscle Cell Plasticity and Fate. Arterioscler Thromb Vasc Biol 2016; 36:1928-36. [PMID: 27470512 DOI: 10.1161/atvbaha.116.308017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 07/11/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Vascular smooth muscle cells (VSMCs) modulate their phenotype between synthetic and contractile states in response to environmental changes; this modulation plays a crucial role in the pathogenesis of restenosis and atherosclerosis. Here, we identified fibroblast growth factor 12 (FGF12) as a novel key regulator of the VSMC phenotype switch. APPROACH AND RESULTS Using murine models and human specimens, we found that FGF12 was highly expressed in contractile VSMCs of normal vessel walls but was downregulated in synthetic VSMCs from injured and atherosclerotic vessels. In human VSMCs, FGF12 expression was inhibited at the transcriptional level by platelet-derived growth factor-BB. Gain- and loss-of-function experiments showed that FGF12 was both necessary and sufficient for inducing and maintaining the quiescent and contractile phenotypes of VSMCs. FGF12 inhibited cell proliferation through the p53 pathway and upregulated the key factors involved in VSMC lineage differentiation, such as myocardin and serum response factor. Such FGF12-induced phenotypic change was mediated by the p38 MAPK (mitogen-activated protein kinase) pathway. Moreover, FGF12 promoted the differentiation of mouse embryonic stem cells and the transdifferentiation of human dermal fibroblasts into SMC-like cells. Furthermore, adenoviral infection of FGF12 substantially decreased neointima hyperplasia in a rat carotid artery injury model. CONCLUSIONS In general, FGF family members induce a synthetic VSMC phenotype. Interestingly, the present study showed the unanticipated finding that FGF12 belonging to FGF family, strongly induced the quiescent and contractile VSMC phenotypes and directly promoted VSMC lineage differentiation. These novel findings suggested that FGF12 could be a new therapeutic target for treating restenosis and atherosclerosis.
Collapse
Affiliation(s)
- Sun-Hwa Song
- From the College of Pharmacy (S.-H.S., K.K., E.-K.J., W.S.), Department of Ophthalmology, College of Medicine (J.T.K.), Chung-Ang University, Seoul, Korea; Division of Vascular Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (Y.-W.K.); Division of Cardiovascular and Rare Disease, Center for Biomedical Sciences, Korea National Institute of Health, Osong, Cheongju, Chungbuk, Korea (J.-S.K.); Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Korea (S.S.B.); College of Pharmacy, Yonsei University, Incheon, Korea (J.-H.S.); STEMORE Co. Ltd., Incheon, Korea (J.-H.S.); and College of Pharmacy, Ajou University, Suwon, Korea (S.G.P.)
| | - Kyungjong Kim
- From the College of Pharmacy (S.-H.S., K.K., E.-K.J., W.S.), Department of Ophthalmology, College of Medicine (J.T.K.), Chung-Ang University, Seoul, Korea; Division of Vascular Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (Y.-W.K.); Division of Cardiovascular and Rare Disease, Center for Biomedical Sciences, Korea National Institute of Health, Osong, Cheongju, Chungbuk, Korea (J.-S.K.); Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Korea (S.S.B.); College of Pharmacy, Yonsei University, Incheon, Korea (J.-H.S.); STEMORE Co. Ltd., Incheon, Korea (J.-H.S.); and College of Pharmacy, Ajou University, Suwon, Korea (S.G.P.)
| | - Eun-Kyung Jo
- From the College of Pharmacy (S.-H.S., K.K., E.-K.J., W.S.), Department of Ophthalmology, College of Medicine (J.T.K.), Chung-Ang University, Seoul, Korea; Division of Vascular Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (Y.-W.K.); Division of Cardiovascular and Rare Disease, Center for Biomedical Sciences, Korea National Institute of Health, Osong, Cheongju, Chungbuk, Korea (J.-S.K.); Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Korea (S.S.B.); College of Pharmacy, Yonsei University, Incheon, Korea (J.-H.S.); STEMORE Co. Ltd., Incheon, Korea (J.-H.S.); and College of Pharmacy, Ajou University, Suwon, Korea (S.G.P.)
| | - Young-Wook Kim
- From the College of Pharmacy (S.-H.S., K.K., E.-K.J., W.S.), Department of Ophthalmology, College of Medicine (J.T.K.), Chung-Ang University, Seoul, Korea; Division of Vascular Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (Y.-W.K.); Division of Cardiovascular and Rare Disease, Center for Biomedical Sciences, Korea National Institute of Health, Osong, Cheongju, Chungbuk, Korea (J.-S.K.); Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Korea (S.S.B.); College of Pharmacy, Yonsei University, Incheon, Korea (J.-H.S.); STEMORE Co. Ltd., Incheon, Korea (J.-H.S.); and College of Pharmacy, Ajou University, Suwon, Korea (S.G.P.)
| | - Jin-Sook Kwon
- From the College of Pharmacy (S.-H.S., K.K., E.-K.J., W.S.), Department of Ophthalmology, College of Medicine (J.T.K.), Chung-Ang University, Seoul, Korea; Division of Vascular Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (Y.-W.K.); Division of Cardiovascular and Rare Disease, Center for Biomedical Sciences, Korea National Institute of Health, Osong, Cheongju, Chungbuk, Korea (J.-S.K.); Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Korea (S.S.B.); College of Pharmacy, Yonsei University, Incheon, Korea (J.-H.S.); STEMORE Co. Ltd., Incheon, Korea (J.-H.S.); and College of Pharmacy, Ajou University, Suwon, Korea (S.G.P.)
| | - Sun Sik Bae
- From the College of Pharmacy (S.-H.S., K.K., E.-K.J., W.S.), Department of Ophthalmology, College of Medicine (J.T.K.), Chung-Ang University, Seoul, Korea; Division of Vascular Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (Y.-W.K.); Division of Cardiovascular and Rare Disease, Center for Biomedical Sciences, Korea National Institute of Health, Osong, Cheongju, Chungbuk, Korea (J.-S.K.); Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Korea (S.S.B.); College of Pharmacy, Yonsei University, Incheon, Korea (J.-H.S.); STEMORE Co. Ltd., Incheon, Korea (J.-H.S.); and College of Pharmacy, Ajou University, Suwon, Korea (S.G.P.)
| | - Jong-Hyuk Sung
- From the College of Pharmacy (S.-H.S., K.K., E.-K.J., W.S.), Department of Ophthalmology, College of Medicine (J.T.K.), Chung-Ang University, Seoul, Korea; Division of Vascular Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (Y.-W.K.); Division of Cardiovascular and Rare Disease, Center for Biomedical Sciences, Korea National Institute of Health, Osong, Cheongju, Chungbuk, Korea (J.-S.K.); Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Korea (S.S.B.); College of Pharmacy, Yonsei University, Incheon, Korea (J.-H.S.); STEMORE Co. Ltd., Incheon, Korea (J.-H.S.); and College of Pharmacy, Ajou University, Suwon, Korea (S.G.P.)
| | - Sang Gyu Park
- From the College of Pharmacy (S.-H.S., K.K., E.-K.J., W.S.), Department of Ophthalmology, College of Medicine (J.T.K.), Chung-Ang University, Seoul, Korea; Division of Vascular Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (Y.-W.K.); Division of Cardiovascular and Rare Disease, Center for Biomedical Sciences, Korea National Institute of Health, Osong, Cheongju, Chungbuk, Korea (J.-S.K.); Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Korea (S.S.B.); College of Pharmacy, Yonsei University, Incheon, Korea (J.-H.S.); STEMORE Co. Ltd., Incheon, Korea (J.-H.S.); and College of Pharmacy, Ajou University, Suwon, Korea (S.G.P.)
| | - Jee Taek Kim
- From the College of Pharmacy (S.-H.S., K.K., E.-K.J., W.S.), Department of Ophthalmology, College of Medicine (J.T.K.), Chung-Ang University, Seoul, Korea; Division of Vascular Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (Y.-W.K.); Division of Cardiovascular and Rare Disease, Center for Biomedical Sciences, Korea National Institute of Health, Osong, Cheongju, Chungbuk, Korea (J.-S.K.); Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Korea (S.S.B.); College of Pharmacy, Yonsei University, Incheon, Korea (J.-H.S.); STEMORE Co. Ltd., Incheon, Korea (J.-H.S.); and College of Pharmacy, Ajou University, Suwon, Korea (S.G.P.)
| | - Wonhee Suh
- From the College of Pharmacy (S.-H.S., K.K., E.-K.J., W.S.), Department of Ophthalmology, College of Medicine (J.T.K.), Chung-Ang University, Seoul, Korea; Division of Vascular Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (Y.-W.K.); Division of Cardiovascular and Rare Disease, Center for Biomedical Sciences, Korea National Institute of Health, Osong, Cheongju, Chungbuk, Korea (J.-S.K.); Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Korea (S.S.B.); College of Pharmacy, Yonsei University, Incheon, Korea (J.-H.S.); STEMORE Co. Ltd., Incheon, Korea (J.-H.S.); and College of Pharmacy, Ajou University, Suwon, Korea (S.G.P.).
| |
Collapse
|
23
|
Furumatsu T, Maehara A, Ozaki T. ROCK inhibition stimulates SOX9/Smad3-dependent COL2A1 expression in inner meniscus cells. J Orthop Sci 2016; 21:524-529. [PMID: 27113646 DOI: 10.1016/j.jos.2016.02.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 01/07/2016] [Accepted: 03/27/2016] [Indexed: 02/09/2023]
Abstract
BACKGROUND Proper functioning of the meniscus depends on the composition and organization of its fibrocartilaginous extracellular matrix. We previously demonstrated that the avascular inner meniscus has a more chondrocytic phenotype compared with the outer meniscus. Inhibition of the Rho family GTPase ROCK, the major regulator of the actin cytoskeleton, stimulates the chondrogenic transcription factor Sry-type HMG box (SOX) 9-dependent α1(II) collagen (COL2A1) expression in inner meniscus cells. However, the crosstalk between ROCK inhibition, SOX9, and other transcription modulators on COL2A1 upregulation remains unclear in meniscus cells. The aim of this study was to investigate the role of SOX9-related transcriptional complex on COL2A1 expression under the inhibition of ROCK in human meniscus cells. METHODS Human inner and outer meniscus cells were prepared from macroscopically intact lateral menisci. Cells were cultured in the presence or absence of ROCK inhibitor (ROCKi, Y27632). Gene expression, collagen synthesis, and nuclear translocation of SOX9 and Smad2/3 were analyzed. RESULTS Treatment of ROCKi increased the ratio of type I/II collagen double positive cells derived from the inner meniscus. In real-time PCR analyses, expression of SOX9 and COL2A1 genes was stimulated by ROCKi treatment in inner meniscus cells. ROCKi treatment also induced nuclear translocation of SOX9 and phosphorylated Smad2/3 in immunohistological analyses. Complex formation between SOX9 and Smad3 was increased by ROCKi treatment in inner meniscus cells. Chromatin immunoprecipitation analyses revealed that association between SOX9/Smad3 transcriptional complex with the COL2A1 enhancer region was increased by ROCKi treatment. CONCLUSIONS This study demonstrated that ROCK inhibition stimulated SOX9/Smad3-dependent COL2A1 expression through the immediate nuclear translocation of Smad3 in inner meniscus cells. Our results suggest that ROCK inhibition can stimulates type II collagen synthesis through the cooperative activation of Smad3 in inner meniscus cells. ROCKi treatment may be useful to promote the fibrochondrocytic healing of the injured inner meniscus.
Collapse
Affiliation(s)
- Takayuki Furumatsu
- Department of Orthopaedic Surgery, Okayama University Graduate School, 2-5-1 Shikata-cho, Kita-ku, Okayama, Japan.
| | - Ami Maehara
- Department of Orthopaedic Surgery, Okayama University Graduate School, 2-5-1 Shikata-cho, Kita-ku, Okayama, Japan
| | - Toshifumi Ozaki
- Department of Orthopaedic Surgery, Okayama University Graduate School, 2-5-1 Shikata-cho, Kita-ku, Okayama, Japan
| |
Collapse
|
24
|
Angelopoulos I, Southern P, Pankhurst QA, Day RM. Superparamagnetic iron oxide nanoparticles regulate smooth muscle cell phenotype. J Biomed Mater Res A 2016; 104:2412-9. [PMID: 27176658 PMCID: PMC5006844 DOI: 10.1002/jbm.a.35780] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/21/2016] [Accepted: 05/11/2016] [Indexed: 01/12/2023]
Abstract
Superparamagnetic iron oxide nanoparticles (SPION) are used for an increasing range of biomedical applications, from imaging to mechanical actuation of cells and tissue. The aim of this study was to investigate the loading of smooth muscle cells (SMC) with SPION and to explore what effect this has on the phenotype of the cells. Adherent human SMC were loaded with ∼17 pg of unconjugated, negatively charged, 50 nm SPION. Clusters of the internalized SPION particles were held in discrete cytoplasmic vesicles. Internalized SPION did not cause any change in cell morphology, proliferation, metabolic activity, or staining pattern of actin and calponin, two of the muscle contractile proteins involved in force generation. However, internalized SPION inhibited the increased gene expression of actin and calponin normally observed when cells are incubated under differentiation conditions. The observed change in the control of gene expression of muscle contractile apparatus by SPION has not previously been described. This finding could offer novel approaches for regulating the phenotype of SMC and warrants further investigation. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 2412–2419, 2016.
Collapse
Affiliation(s)
- Ioannis Angelopoulos
- Applied Biomedical Engineering Group, Division of Medicine, University College London, WC1E 6DD, UK
| | - Paul Southern
- UCL Institute of Biomedical Engineering, University College London, London, WC1E 6BT, UK
| | - Quentin A Pankhurst
- UCL Institute of Biomedical Engineering, University College London, London, WC1E 6BT, UK
| | - Richard M Day
- Applied Biomedical Engineering Group, Division of Medicine, University College London, WC1E 6DD, UK
| |
Collapse
|
25
|
Almalki SG, Agrawal DK. Key transcription factors in the differentiation of mesenchymal stem cells. Differentiation 2016; 92:41-51. [PMID: 27012163 DOI: 10.1016/j.diff.2016.02.005] [Citation(s) in RCA: 275] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/15/2016] [Accepted: 02/25/2016] [Indexed: 11/15/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that represent a promising source for regenerative medicine. MSCs are capable of osteogenic, chondrogenic, adipogenic and myogenic differentiation. Efficacy of differentiated MSCs to regenerate cells in the injured tissues requires the ability to maintain the differentiation toward the desired cell fate. Since MSCs represent an attractive source for autologous transplantation, cellular and molecular signaling pathways and micro-environmental changes have been studied in order to understand the role of cytokines, chemokines, and transcription factors on the differentiation of MSCs. The differentiation of MSC into a mesenchymal lineage is genetically manipulated and promoted by specific transcription factors associated with a particular cell lineage. Recent studies have explored the integration of transcription factors, including Runx2, Sox9, PPARγ, MyoD, GATA4, and GATA6 in the differentiation of MSCs. Therefore, the overexpression of a single transcription factor in MSCs may promote trans-differentiation into specific cell lineage, which can be used for treatment of some diseases. In this review, we critically discussed and evaluated the role of transcription factors and related signaling pathways that affect the differentiation of MSCs toward adipocytes, chondrocytes, osteocytes, skeletal muscle cells, cardiomyocytes, and smooth muscle cells.
Collapse
Affiliation(s)
- Sami G Almalki
- Departments of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, USA
| | - Devendra K Agrawal
- Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, USA.
| |
Collapse
|
26
|
Liu H, Liu A, Shi C, Li B. Curcumin suppresses transforming growth factor-β1-induced cardiac fibroblast differentiation via inhibition of Smad-2 and p38 MAPK signaling pathways. Exp Ther Med 2016; 11:998-1004. [PMID: 26998027 DOI: 10.3892/etm.2016.2969] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 12/10/2015] [Indexed: 02/06/2023] Open
Abstract
The differentiation of cardiac fibroblasts (CFs) into myofibroblasts and the subsequent deposition of the extracellular matrix is associated with myocardial fibrosis following various types of myocardial injury. In the present study, the effect of curcumin, which is a pharmacologically-safe natural compound from the Curcuma longa herb, on transforming growth factor (TGF)-β1-induced CFs was investigated, and the underlying molecular mechanisms were examined. The expression levels of α-smooth muscle actin (SMA) stress fibers were investigated using western blotting and immunofluorescence in cultured neonatal rat CFs. Protein and mRNA expression levels of α-SMA and collagen type I (ColI) were determined by western blotting and reverse transcription-quantitative polymerase chain reaction. In addition, the activation of Smad2 and p38 was examined using western blotting. Curcumin, SB431542 (a TGF-βR-Smad2 inhibitor) and SB203580 (a p38 inhibitor) were used to inhibit the stimulation by TGF-β1. The results demonstrated that the TGF-β1-induced expression of α-SMA and ColI was suppressed by curcumin at the mRNA and protein levels, while SB431542 and SB203580 induced similar effects. Furthermore, phosphorylated Smad-2 and p38 were upregulated in TGF-β1-induced CFs, and these effects were substantially inhibited by curcumin administration. In conclusion, the results of the present study demonstrated that treatment with curcumin effectively suppresses TGF-β1-induced CF differentiation via Smad-2 and p38 signaling pathways. Thus, curcumin may be a potential therapeutic agent for the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Huzi Liu
- Department of Cardiac Surgery, Shanxi Cardiovascular Hospital, Taiyuan, Shanxi 030024, P.R. China
| | - Aijun Liu
- Pediatric Heart Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Chunli Shi
- Outpatient Department, Shanxi Cardiovascular Hospital, Taiyuan, Shanxi 030024, P.R. China
| | - Bao Li
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan, Shanxi 030024, P.R. China
| |
Collapse
|
27
|
PRK1/PKN1 controls migration and metastasis of androgen-independent prostate cancer cells. Oncotarget 2015; 5:12646-64. [PMID: 25504435 PMCID: PMC4350344 DOI: 10.18632/oncotarget.2653] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 10/26/2014] [Indexed: 12/20/2022] Open
Abstract
The major threat in prostate cancer is the occurrence of metastases in androgen-independent tumor stage, for which no causative cure is available. Here we show that metastatic behavior of androgen-independent prostate tumor cells requires the protein-kinase-C-related kinase (PRK1/PKN1) in vitro and in vivo. PRK1 regulates cell migration and gene expression through its kinase activity, but does not affect cell proliferation. Transcriptome and interactome analyses uncover that PRK1 regulates expression of migration-relevant genes by interacting with the scaffold protein sperm-associated antigen 9 (SPAG9/JIP4). SPAG9 and PRK1 colocalize in human cancer tissue and are required for p38-phosphorylation and cell migration. Accordingly, depletion of either ETS domain-containing protein Elk-1 (ELK1), an effector of p38-signalling or p38 depletion hinders cell migration and changes expression of migration-relevant genes as observed upon PRK1-depletion. Importantly, a PRK1 inhibitor prevents metastases in mice, showing that the PRK1-pathway is a promising target to hamper prostate cancer metastases in vivo.
Collapse
|
28
|
Moharil J, Lei P, Tian J, Gaile DP, Andreadis ST. Lentivirus Live Cell Array for Quantitative Assessment of Gene and Pathway Activation during Myogenic Differentiation of Mesenchymal Stem Cells. PLoS One 2015; 10:e0141365. [PMID: 26505747 PMCID: PMC4624764 DOI: 10.1371/journal.pone.0141365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/06/2015] [Indexed: 11/19/2022] Open
Abstract
Stem cell differentiation involves multiple cascades of transcriptional regulation that govern the cell fate. To study the real-time dynamics of this complex process, quantitative and high throughput live cell assays are required. Herein, we developed a lentiviral library of promoters and transcription factor binding sites to quantitatively capture the gene expression dynamics over a period of several days during myogenic differentiation of human mesenchymal stem cells (MSCs) harvested from two different anatomic locations, bone marrow and hair follicle. Our results enabled us to monitor the sequential activation of signaling pathways and myogenic gene promoters at various stages of differentiation. In conjunction with chemical inhibitors, the lentiviral array (LVA) results also revealed the relative contribution of key signaling pathways that regulate the myogenic differentiation. Our study demonstrates the potential of LVA to monitor the dynamics of gene and pathway activation during MSC differentiation as well as serve as a platform for discovery of novel molecules, genes and pathways that promote or inhibit complex biological processes.
Collapse
Affiliation(s)
- Janhavi Moharil
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, 908 Furnas Hall, Amherst, NY 14260–4200, United States of America
- Department of Biostatistics, University at Buffalo, State University of New York, Kimball, Buffalo, NY 14214–3000, United States of America
| | - Pedro Lei
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, 908 Furnas Hall, Amherst, NY 14260–4200, United States of America
| | - Jun Tian
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, 908 Furnas Hall, Amherst, NY 14260–4200, United States of America
| | - Daniel P. Gaile
- Department of Biostatistics, University at Buffalo, State University of New York, Kimball, Buffalo, NY 14214–3000, United States of America
| | - Stelios T. Andreadis
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, 908 Furnas Hall, Amherst, NY 14260–4200, United States of America
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260–4200, United States of America
- Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14203, United States of America
- * E-mail:
| |
Collapse
|
29
|
Shi N, Chen SY. Smooth Muscle Cell Differentiation: Model Systems, Regulatory Mechanisms, and Vascular Diseases. J Cell Physiol 2015; 231:777-87. [DOI: 10.1002/jcp.25208] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 09/29/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Ning Shi
- Department of Physiology and Pharmacology; University of Georgia; Athens Georgia
| | - Shi-You Chen
- Department of Physiology and Pharmacology; University of Georgia; Athens Georgia
| |
Collapse
|
30
|
Koeck I, Burkhard FC, Monastyrskaya K. Activation of common signaling pathways during remodeling of the heart and the bladder. Biochem Pharmacol 2015; 102:7-19. [PMID: 26390804 DOI: 10.1016/j.bcp.2015.09.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/14/2015] [Indexed: 12/12/2022]
Abstract
The heart and the urinary bladder are hollow muscular organs, which can be afflicted by pressure overload injury due to pathological conditions such as hypertension and bladder outlet obstruction. This increased outflow resistance induces hypertrophy, marked by dramatic changes in the organs' phenotype and function. The end result in both the heart and the bladder can be acute organ failure due to advanced fibrosis and the subsequent loss of contractility. There is emerging evidence that microRNAs (miRNAs) play an important role in the pathogenesis of heart failure and bladder dysfunction. MiRNAs are endogenous non-coding single-stranded RNAs, which regulate gene expression and control adaptive and maladaptive organ remodeling processes. This Review summarizes the current knowledge of molecular alterations in the heart and the bladder and highlights common signaling pathways and regulatory events. The miRNA expression analysis and experimental target validation done in the heart provide a valuable source of information for investigators working on the bladder and other organs undergoing the process of fibrotic remodeling. Aberrantly expressed miRNA are amendable to pharmacological manipulation, offering an opportunity for development of new therapies for cardiac and bladder hypertrophy and failure.
Collapse
Affiliation(s)
- Ivonne Koeck
- Urology Research Laboratory, Department Clinical Research, University of Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | | | - Katia Monastyrskaya
- Urology Research Laboratory, Department Clinical Research, University of Bern, Switzerland; Department of Urology, University Hospital, Bern, Switzerland.
| |
Collapse
|
31
|
Shi N, Guo X, Chen SY. Olfactomedin 2, a novel regulator for transforming growth factor-β-induced smooth muscle differentiation of human embryonic stem cell-derived mesenchymal cells. Mol Biol Cell 2014; 25:4106-14. [PMID: 25298399 PMCID: PMC4263453 DOI: 10.1091/mbc.e14-08-1255] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Smooth muscle plays important roles in vascular development. Study of smooth muscle differentiation of human embryonic stem cell–derived mesenchymal cells identifies olfactomedin 2 as a novel regulator. Olfactomedin 2 regulates smooth muscle gene transcription by empowering serum response factor binding to the CArG box in smooth muscle gene promoters. Transforming growth factor-β (TGF-β) plays an important role in smooth muscle (SM) differentiation, but the downstream target genes regulating the differentiation process remain largely unknown. In this study, we identified olfactomedin 2 (Olfm2) as a novel regulator mediating SM differentiation. Olfm2 was induced during TGF-β–induced SM differentiation of human embryonic stem cell–derived mesenchymal cells. Olfm2 knockdown suppressed TGF-β–induced expression of SM markers, including SM α-actin, SM22α, and SM myosin heavy chain, whereas Olfm2 overexpression promoted the SM marker expression. TGF-β induced Olfm2 nuclear accumulation, suggesting that Olfm2 may be involved in transcriptional activation of SM markers. Indeed, Olfm2 regulated SM marker expression and promoter activity in a serum response factor (SRF)/CArG box–dependent manner. Olfm2 physically interacted with SRF without affecting SRF-myocardin interaction. Olfm2-SRF interaction promoted the dissociation of SRF from HERP1, a transcriptional repressor. Olfm2 also inhibited HERP1 expression. Moreover, blockade of Olfm2 expression inhibited TGF-β–induced SRF binding to SM gene promoters in a chromatin setting, whereas overexpression of Olfm2 dose dependently enhanced SRF binding. These results demonstrate that Olfm2 mediates TGF-β–induced SM gene transcription by empowering SRF binding to CArG box in SM gene promoters.
Collapse
Affiliation(s)
- Ning Shi
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602
| | - Xia Guo
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602
| | - Shi-You Chen
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602
| |
Collapse
|
32
|
Ye GJC, Aratyn-Schaus Y, Nesmith AP, Pasqualini FS, Alford PW, Parker KK. The contractile strength of vascular smooth muscle myocytes is shape dependent. Integr Biol (Camb) 2014; 6:152-63. [PMID: 24406783 DOI: 10.1039/c3ib40230d] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Vascular smooth muscle cells in muscular arteries are more elongated than those in elastic arteries. Previously, we reported changes in the contractility of engineered vascular smooth muscle tissue that appeared to be correlated with the shape of the constituent cells, supporting the commonly held belief that elongated muscle geometry may allow for the better contractile tone modulation required in response to changes in blood flow and pressure. To test this hypothesis more rigorously, we developed an in vitro model by engineering human vascular smooth muscle cells to take on the same shapes as those seen in elastic and muscular arteries and measured their contraction during stimulation with endothelin-1. We found that in the engineered cells, actin alignment and nuclear eccentricity increased as the shape of the cell elongated. Smooth muscle cells with elongated shapes exhibited lower contractile strength but greater percentage increase in contraction after endothelin-1 stimulation. We analysed the relationship between smooth muscle contractility and subcellular architecture and found that changes in contractility were correlated with actin alignment and nuclear shape. These results suggest that elongated smooth muscle cells facilitate muscular artery tone modulation by increasing its dynamic contractile range.
Collapse
Affiliation(s)
- George J C Ye
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering and the School of Engineering and Applied Sciences, Harvard University, 29 Oxford St, Pierce Hall 321, Cambridge, MA 02138, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Nakamoto T, Wang X, Kawazoe N, Chen G. Influence of micropattern width on differentiation of human mesenchymal stem cells to vascular smooth muscle cells. Colloids Surf B Biointerfaces 2014; 122:316-323. [PMID: 25064482 DOI: 10.1016/j.colsurfb.2014.06.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Revised: 05/27/2014] [Accepted: 06/04/2014] [Indexed: 12/31/2022]
Abstract
In recent years, various approaches have been taken to generate functional muscle tissue by tissue engineering. However, in vitro methods to generate smooth muscle with physiologically aligned structure remains limited. In order to mimic the in vivo highly organized structure of smooth muscle cells, we used micropatterning technology for engineering parallel aligned cells. In this study, a gradient micropattern of different width of cell-adhesive polystyrene stripes (5, 10, 20, 40, 60, 80, 100, 200, 400, 600, 800 and 1000μm) was prepared and the effects of micropattern width on human mesenchymal stem cells (hMSCs) orientation, morphology and smooth muscle cell differentiation were investigated. The width of micropattern stripes showed obvious effect on cell orientation, morphology and smooth muscle cell differentiation. The cells showed higher degree of orientation when the micropattern stripes became narrower. Higher expression of calponin and smooth muscle actin was observed among the narrow micropatterns ranging from 200μm to 20μm, compared to the non-patterned area and wide micropattern areas which showed similar levels of expression.
Collapse
Affiliation(s)
- Tomoko Nakamoto
- Tissue Regeneration Materials Unit, International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan.
| | - Xinlong Wang
- Tissue Regeneration Materials Unit, International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Department of Materials Science and Engineering, Graduate School of Pure and Applied Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan.
| | - Naoki Kawazoe
- Tissue Regeneration Materials Unit, International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan.
| | - Guoping Chen
- Tissue Regeneration Materials Unit, International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Department of Materials Science and Engineering, Graduate School of Pure and Applied Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan.
| |
Collapse
|
34
|
Hu B, Song JT, Qu HY, Bi CL, Huang XZ, Liu XX, Zhang M. Mechanical stretch suppresses microRNA-145 expression by activating extracellular signal-regulated kinase 1/2 and upregulating angiotensin-converting enzyme to alter vascular smooth muscle cell phenotype. PLoS One 2014; 9:e96338. [PMID: 24848371 PMCID: PMC4029552 DOI: 10.1371/journal.pone.0096338] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Accepted: 04/04/2014] [Indexed: 11/18/2022] Open
Abstract
Phenotype modulation of vascular smooth muscle cells (VSMCs) plays an important role in the pathogenesis of various vascular diseases, including hypertension and atherosclerosis. Several microRNAs (miRNAs) were found involved in regulating the VSMC phenotype with platelet-derived growth factor (PDGF) treatment, but the role of miRNAs in the mechanical stretch-altered VSMC phenotype is not clear. Here, we identified miR-145 as a major miRNA contributing to stretch-altered VSMC phenotype by miRNA array, quantitative RT-PCR and gain- and loss-of-function methods. Our data demonstrated that 16% stretch suppressed miR-145 expression, with reduced expression of contractile markers of VSMCs cultured on collagenI; overexpression of miR-145 could partially recover the expression in stretched cells. Serum response factor (SRF), myocardin, and Kruppel-like factor 4 (KLF4) are major regulators of the VSMC phenotype. The effect of stretch on myocardin and KLF4 protein expression was altered by miR-145 mimics, but SRF expression was not affected. In addition, stretch-activated extracellular signal-regulated kinase 1/2 (ERK1/2) and up-regulated angiotensin-converting enzyme (ACE) were confirmed to be responsible for the inhibition of miR-145 expression. Mechanical stretch inhibits miR-145 expression by activating the ERK1/2 signaling pathway and promoting ACE expression, thus modulating the VSMC phenotype.
Collapse
Affiliation(s)
- Bo Hu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Jian tao Song
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Hai yan Qu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Chen long Bi
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Xiao zhen Huang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Xin xin Liu
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Mei Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Department of Cardiology, Qilu Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
- * E-mail:
| |
Collapse
|
35
|
Penke LRK, Huang SK, White ES, Peters-Golden M. Prostaglandin E2 inhibits α-smooth muscle actin transcription during myofibroblast differentiation via distinct mechanisms of modulation of serum response factor and myocardin-related transcription factor-A. J Biol Chem 2014; 289:17151-62. [PMID: 24802754 DOI: 10.1074/jbc.m114.558130] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Differentiation of lung fibroblasts into contractile protein-expressing myofibroblasts by transforming growth factor-β1 (TGF-β1) is a critical event in the pathogenesis of pulmonary fibrosis. Transcription of the contractile protein α-smooth muscle actin (α-SMA) is mediated by the transcription factor serum-response factor (SRF) along with its co-activator, myocardin-related transcription factor-A (MRTF-A). The endogenous lipid mediator prostaglandin E2 (PGE2) exerts anti-fibrotic effects, including the inhibition of myofibroblast differentiation. However, the mechanism by which PGE2 inhibits α-SMA expression is incompletely understood. Here, we show in normal lung fibroblasts that PGE2 reduced the nuclear accumulation of MRTF-A·SRF complexes and consequently inhibited α-SMA promoter activation. It did so both by independently inhibiting SRF gene expression and nuclear import of MRTF-A. We identified that p38 MAPK is critical for TGF-β1-induced SRF gene expression and that PGE2 inhibition of SRF expression is associated with its ability to inhibit p38 activation. Its inhibition of MRTF-A import occurs via activation of cofilin 1 and inactivation of vasodilator-stimulated phosphoprotein. Similar effects of PGE2 on SRF gene expression were observed in fibroblasts from the lungs of patients with idiopathic pulmonary fibrosis. Thus, PGE2 is the first substance described to prevent myofibroblast differentiation by disrupting, via distinct mechanisms, the actions of both SRF and MRTF-A.
Collapse
Affiliation(s)
- Loka R K Penke
- From the Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Steven K Huang
- From the Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Eric S White
- From the Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Marc Peters-Golden
- From the Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109
| |
Collapse
|
36
|
Wu ML, Chen CH, Lin YT, Jheng YJ, Ho YC, Yang LT, Chen L, Layne MD, Yet SF. Divergent signaling pathways cooperatively regulate TGFβ induction of cysteine-rich protein 2 in vascular smooth muscle cells. Cell Commun Signal 2014; 12:22. [PMID: 24674138 PMCID: PMC3973006 DOI: 10.1186/1478-811x-12-22] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 03/23/2014] [Indexed: 01/31/2023] Open
Abstract
Background Vascular smooth muscle cells (VSMCs) of the arterial wall play a critical role in the development of occlusive vascular diseases. Cysteine-rich protein 2 (CRP2) is a VSMC-expressed LIM-only protein, which functionally limits VSMC migration and protects against pathological vascular remodeling. The multifunctional cytokine TGFβ has been implicated to play a role in the pathogenesis of atherosclerosis through numerous downstream signaling pathways. We showed previously that TGFβ upregulates CRP2 expression; however, the detailed signaling mechanisms remain unclear. Results TGFβ treatment of VSMCs activated both Smad2/3 and ATF2 phosphorylation. Individually knocking down Smad2/3 or ATF2 pathways with siRNA impaired the TGFβ induction of CRP2, indicating that both contribute to CRP2 expression. Inhibiting TβRI kinase activity by SB431542 or TβRI knockdown abolished Smad2/3 phosphorylation but did not alter ATF2 phosphorylation, indicating while Smad2/3 phosphorylation was TβRI-dependent ATF2 phosphorylation was independent of TβRI. Inhibiting Src kinase activity by SU6656 suppressed TGFβ-induced RhoA and ATF2 activation but not Smad2 phosphorylation. Blocking ROCK activity, the major downstream target of RhoA, abolished ATF2 phosphorylation and CRP2 induction but not Smad2 phosphorylation. Furthermore, JNK inhibition with SP600125 reduced TGFβ-induced ATF2 (but not Smad2) phosphorylation and CRP2 protein expression while ROCK inhibition blocked JNK activation. These results indicate that downstream of TβRII, Src family kinase-RhoA-ROCK-JNK signaling pathway mediates TβRI-independent ATF2 activation. Promoter analysis revealed that the TGFβ induction of CRP2 was mediated through the CRE and SBE promoter elements that were located in close proximity. Conclusions Our results demonstrate that two signaling pathways downstream of TGFβ converge on the CRE and SBE sites of the Csrp2 promoter to cooperatively control CRP2 induction in VSMCs, which represents a previously unrecognized mechanism of VSMC gene induction by TGFβ.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Shaw-Fang Yet
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan.
| |
Collapse
|
37
|
Gao Y, Bayless KJ, Li Q. TGFBR1 is required for mouse myometrial development. Mol Endocrinol 2014; 28:380-94. [PMID: 24506537 PMCID: PMC3938542 DOI: 10.1210/me.2013-1284] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 01/27/2014] [Indexed: 12/22/2022] Open
Abstract
The smooth muscle layer of the uterus (ie, myometrium) is critical for a successful pregnancy and labor. We have shown that the conditional deletion of TGFβ type 1 receptor (TGFBR1) in the female reproductive tract leads to remarkable smooth muscle defects. This study was aimed at defining the cellular and molecular basis of the myometrial defects. We found that TGFBR1 is required for myometrial configuration and formation during early postnatal uterine development. Despite the well-established role of TGFβ signaling in vascular smooth muscle cell differentiation, the majority of smooth muscle genes were expressed in Tgfbr1 conditional knockout (cKO) uteri at similar levels as controls during postnatal uterine development, coinciding with the presence but abnormal distribution of proteins for select smooth muscle markers. Importantly, the uteri of these mice had impaired synthesis of key extracellular matrix proteins and dysregulated expression of platelet-derived growth factors. Furthermore, platelet-derived growth factors induced the migration of uterine stromal cells from both control and Tgfbr1 cKO mice in vitro. Our results suggest that the myometrial defects in Tgfbr1 cKO mice may not directly arise from an intrinsic deficiency in uterine smooth muscle cell differentiation but are linked to the impaired production of key extracellular matrix components and abnormal uterine cell migration during a critical time window of postnatal uterine development. These findings will potentially aid in the design of novel therapies for reproductive disorders associated with myometrial defects.
Collapse
Affiliation(s)
- Yang Gao
- Department of Veterinary Integrative Biosciences (Y.G., Q.L.), College of Veterinary Medicine and Biomedical Sciences, and Department of Molecular and Cellular Medicine (K.J.B.), Texas A&M Health Science Center, Texas A&M University, College Station, Texas 77843
| | | | | |
Collapse
|
38
|
Shi N, Chen SY. Mechanisms simultaneously regulate smooth muscle proliferation and differentiation. J Biomed Res 2013; 28:40-6. [PMID: 24474962 PMCID: PMC3904173 DOI: 10.7555/jbr.28.20130130] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 11/13/2013] [Indexed: 01/01/2023] Open
Abstract
Vascular smooth muscle cell (VSMC) differentiation and proliferation are two important physiological processes during vascular development. The phenotypic alteration from differentiated to proliferative VSMC contributes to the development of several major cardiovascular diseases including atherosclerosis, hypertension, restenosis after angioplasty or bypass, diabetic vascular complications, and transplantation arteriopathy. Since the VSMC phenotype in these pathological conditions resembles that of developing VSMC during embryonic development, understanding of the molecular mechanisms that control VSMC differentiation will provide fundamental insights into the pathological processes of these cardiovascular diseases. Although VSMC differentiation is usually accompanied by an irreversible cell cycle exit, VSMC proliferation and differentiation occur concurrently during embryonic development. The molecular mechanisms simultaneously regulating these two processes, however, remain largely unknown. Our recent study demonstrates that cell division cycle 7, a key regulator of cell cycle, promotes both VSMC differentiation and proliferation through different mechanisms during the initial phase of VSMC differentiation. Conversely, Krüppel-like factor 4 appears to be a repressor for both VSMC differentiation and proliferation. This review attempts to highlight the novel role of cell division cycle 7 in TGF-β-induced VSMC differentiation and proliferation. The role of Krüppel-like factor 4 in suppressing these two processes will also be discussed.
Collapse
Affiliation(s)
- Ning Shi
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA 30602, USA
| | - Shi-You Chen
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
39
|
Xu ZC, Zhang Q, Li H. Differentiation of human hair follicle stem cells into endothelial cells induced by vascular endothelial and basic fibroblast growth factors. Mol Med Rep 2013; 9:204-10. [PMID: 24247660 DOI: 10.3892/mmr.2013.1796] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 11/11/2013] [Indexed: 11/06/2022] Open
Abstract
Hair follicle stem cells (HFSCs) possess powerful expansion and multi‑differentiation potential, properties that place them at the forefront of the field of tissue engineering and stem cell‑based therapy. The aim of the present study was to investigate the differentiation of human HFSCs (hHFSCs) into cells of an endothelial lineage. hHFSCs were expanded to the second passage in vitro and then induced by the addition of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) to the culture medium. The expression levels of endothelial cell (EC)‑related markers, including von Willebrand factor (vWF), vascular endothelial cadherin (VE)‑cadherin and cluster of differentiation (CD)31, were detected by immunofluorescence staining, flow cytometric analysis and reverse transcription‑polymerase chain reaction. The hHFSCs expressed vWF, VE‑cadherin and CD31 when exposed to a differentiation medium, similar to the markers expressed by the human umbilical vein ECs. More significantly, differentiated cells were also able to take up low‑density lipoprotein. The data of the present study demonstrated that an efficient strategy may be developed for differentiating hHFSCs into ECs by stimulation with VEGF and bFGF. Thus, hHFSCs represent a novel cell source for vascular tissue engineering and studies regarding the treatment of various forms of ischaemic vascular disease.
Collapse
Affiliation(s)
- Zhi Cheng Xu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, Shanghai 200011, P.R. China
| | | | | |
Collapse
|
40
|
Xu ZC, Zhang Q, Li H. Human hair follicle stem cell differentiation into contractile smooth muscle cells is induced by transforming growth factor-β1 and platelet-derived growth factor BB. Mol Med Rep 2013; 8:1715-21. [PMID: 24084832 DOI: 10.3892/mmr.2013.1707] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Accepted: 09/23/2013] [Indexed: 11/06/2022] Open
Abstract
Smooth muscle cells (SMCs) are important in vascular homeostasis and disease and thus, are critical elements in vascular tissue engineering. Although adult SMCs have been used as seed cells, such mature differentiated cells suffer from limited proliferation potential and cultural senescence, particularly when originating from older donors. By comparison, human hair follicle stem cells (hHFSCs) are a reliable source of stem cells with multi-differentiation potential. The aim of the present study, was to develop an efficient strategy to derive functional SMCs from hHFSCs. hHFSCs were obtained from scalp tissues of healthy adult patients undergoing cosmetic plastic surgery. The hHFSCs were expanded to passage 2 and induced by the administration of transforming growth factor-β1 (TGF-β1) and platelet-derived growth factor BB (PDGF-BB) in combination with culture medium. Expression levels of SMC-related markers, including α-smooth muscle actin (α-SMA), α-calponin and smooth muscle myosin heavy chain (SM-MHC), were detected by immunofluorescence staining, flow cytometry analysis and reverse transcription-polymerase chain reaction (RT-PCR). When exposed to differentiation medium, hHFSCs expressed early, mid and late markers (α-SMA, α-calponin and SM-MHC, respectively) that were similar to the markers expressed by human umbilical artery SMCs. Notably, when entrapped inside a collagen matrix lattice, these SM differentiated cells showed a contractile function. Therefore, the present study developed an efficient strategy for differentiating hHFSCs into contractile SMCs by stimulation with TGF-β1 and PDGF-BB. The high yield of derivation suggests that this strategy facilitates the acquisition of the large numbers of cells that are required for blood vessel engineering and the study of vascular disease pathophysiology.
Collapse
Affiliation(s)
- Zhi Cheng Xu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, P.R. China
| | | | | |
Collapse
|
41
|
Wanjare M, Kusuma S, Gerecht S. Perivascular cells in blood vessel regeneration. Biotechnol J 2013; 8:434-47. [PMID: 23554249 DOI: 10.1002/biot.201200199] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 02/19/2013] [Accepted: 03/05/2013] [Indexed: 12/21/2022]
Abstract
Vascular engineering seeks to design and construct functional blood vessels comprising endothelial cells (ECs) and perivascular cells (PCs), with the ultimate goal of clinical translation. While EC behavior has been extensively investigated, PCs play an equally significant role in the development of novel regenerative strategies, providing functionality and stability to vessels. The two major classes of PCs are vascular smooth muscle cells (vSMCs) and pericytes; vSMCs can be further sub-classified as either contractile or synthetic. The inclusion of these cell types is crucial for successful regeneration of blood vessels. Furthermore, understanding distinctions between vSMCs and pericytes will enable improved therapeutics in a tissue-specific manner. Here we focus on the approaches and challenges facing the use of PCs in vascular regeneration, including their characteristics, stem cell sources, and interactions with ECs. Finally, we discuss biochemical and microRNA (miR) regulators of PC behavior and engineering approaches that mimic various cues affecting PC function.
Collapse
Affiliation(s)
- Maureen Wanjare
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | | | | |
Collapse
|
42
|
Abstract
Vascular smooth muscle cells have attracted considerable interest as a model for a flexible program of gene expression. This cell type arises throughout the embryo body plan via poorly understood signaling cascades that direct the expression of transcription factors and microRNAs which, in turn, orchestrate the activation of contractile genes collectively defining this cell lineage. The discovery of myocardin and its close association with serum response factor has represented a major break-through for the molecular understanding of vascular smooth muscle cell differentiation. Retinoids have been shown to improve the outcome of vessel wall remodeling following injury and have provided further insights into the molecular circuitry that defines the vascular smooth muscle cell phenotype. This review summarizes the progress to date in each of these areas of vascular smooth muscle cell biology.
Collapse
|
43
|
Long X, Cowan SL, Miano JM. Mitogen-activated protein kinase 14 is a novel negative regulatory switch for the vascular smooth muscle cell contractile gene program. Arterioscler Thromb Vasc Biol 2012; 33:378-86. [PMID: 23175675 DOI: 10.1161/atvbaha.112.300645] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Several studies have shown through chemical inhibitors that p38 mitogen-activated protein kinase (MAPK) promotes vascular smooth muscle cell (VSMC) differentiation. Here, we evaluate the effects of knocking down a dominant p38MAPK isoform on VSMC differentiation. METHODS AND RESULTS Knockdown of p38MAPKα (MAPK14) in human coronary artery SMCs unexpectedly increases VSMC differentiation genes, such as miR145, ACTA2, CNN1, LMOD1, and TAGLN, with little change in the expression of serum response factor (SRF) and 2 SRF cofactors, myocardin (MYOCD) and myocardin-related transcription factor A (MKL1). A variety of chemical and biological inhibitors demonstrate a critical role for a RhoA-MKL1-SRF-dependent pathway in mediating these effects. MAPK14 knockdown promotes MKL1 nuclear localization and VSMC marker expression, an effect partially reversed with Y27632; in contrast, MAP2K6 (MKK6) blocks MKL1 nuclear import and VSMC marker expression. Immunostaining and Western blotting of injured mouse carotid arteries reveal elevated MAPK14 (both total and phosphorylated) and reduced VSMC marker expression. CONCLUSIONS Reduced MAPK14 expression evokes unanticipated increases in VSMC contractile genes, suggesting an unrecognized negative regulatory role for MAPK14 signaling in VSMC differentiation.
Collapse
Affiliation(s)
- Xiaochun Long
- Department of Medicine, Aab Cardiovascular Research Institute, Box CVRI, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY 14642, USA.
| | | | | |
Collapse
|
44
|
Mendes LF, Pirraco RP, Szymczyk W, Frias AM, Santos TC, Reis RL, Marques AP. Perivascular-like cells contribute to the stability of the vascular network of osteogenic tissue formed from cell sheet-based constructs. PLoS One 2012; 7:e41051. [PMID: 22829909 PMCID: PMC3400580 DOI: 10.1371/journal.pone.0041051] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 06/16/2012] [Indexed: 12/11/2022] Open
Abstract
In recent years several studies have been supporting the existence of a close relationship in terms of function and progeny between Mesenchymal Stem Cells (MSCs) and Pericytes. This concept has opened new perspectives for the application of MSCs in Tissue Engineering (TE), with special interest for the pre-vascularization of cell dense constructs. In this work, cell sheet technology was used to create a scaffold-free construct composed of osteogenic, endothelial and perivascular-like (CD146+) cells for improved in vivo vessel formation, maturation and stability. The CD146 pericyte-associated phenotype was induced from human bone marrow mesenchymal stem cells (hBMSCs) by the supplementation of standard culture medium with TGF-β1. Co-cultured cell sheets were obtained by culturing perivascular-like (CD146+) cells and human umbilical vein endothelial cells (HUVECs) on an hBMSCs monolayer maintained in osteogenic medium for 7 days. The perivascular-like (CD146+) cells and the HUVECs migrated and organized over the collagen-rich osteogenic cell sheet, suggesting the existence of cross-talk involving the co-cultured cell types. Furthermore the presence of that particular ECM produced by the osteoblastic cells was shown to be the key regulator for the singular observed organization. The osteogenic and angiogenic character of the proposed constructs was assessed in vivo. Immunohistochemistry analysis of the explants revealed the integration of HUVECs with the host vasculature as well as the osteogenic potential of the created construct, by the expression of osteocalcin. Additionally, the analysis of the diameter of human CD146 positive blood vessels showed a higher mean vessel diameter for the co-cultured cell sheet condition, reinforcing the advantage of the proposed model regarding blood vessels maturation and stability and for the in vitro pre-vascularization of TE constructs.
Collapse
Affiliation(s)
- Luís F. Mendes
- 3B’s Research Group–Biomaterials, Biodegradables and Biomimetics, University of Minho, Guimarães, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
| | - Rogério P. Pirraco
- 3B’s Research Group–Biomaterials, Biodegradables and Biomimetics, University of Minho, Guimarães, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
| | - Wojciech Szymczyk
- 3B’s Research Group–Biomaterials, Biodegradables and Biomimetics, University of Minho, Guimarães, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
| | - Ana M. Frias
- 3B’s Research Group–Biomaterials, Biodegradables and Biomimetics, University of Minho, Guimarães, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
| | - Tírcia C. Santos
- 3B’s Research Group–Biomaterials, Biodegradables and Biomimetics, University of Minho, Guimarães, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
| | - Rui L. Reis
- 3B’s Research Group–Biomaterials, Biodegradables and Biomimetics, University of Minho, Guimarães, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
| | - Alexandra P. Marques
- 3B’s Research Group–Biomaterials, Biodegradables and Biomimetics, University of Minho, Guimarães, Portugal
- ICVS/3B’s, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
- * E-mail:
| |
Collapse
|
45
|
Tang Y, Boucher JM, Liaw L. Histone deacetylase activity selectively regulates notch-mediated smooth muscle differentiation in human vascular cells. J Am Heart Assoc 2012; 1:e000901. [PMID: 23130137 PMCID: PMC3487326 DOI: 10.1161/jaha.112.000901] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 05/16/2012] [Indexed: 12/17/2022]
Abstract
Background Histone deacetylases (HDACs) modify smooth muscle cell (SMC) proliferation and affect neointimal lesion formation by regulating cell cycle progression. HDACs might also regulate SMC differentiation, although this is not as well characterized. Methods and Results Notch signaling activates SMC contractile markers and the differentiated phenotype in human aortic SMCs. Using this model, we found that HDAC inhibition antagonized the ability of Notch to increase levels of smooth muscle α-actin, calponin1, smooth muscle 22α, and smooth muscle myosin heavy chain. However, inhibition of HDAC activity did not suppress Notch activation of the HRT target genes. In fact, HDAC inhibition increased activation of the canonical C-promoter binding factor-1 (CBF-1)–mediated Notch pathway, which activates HRT transcription. Although CBF-1–mediated Notch signaling was increased by HDAC inhibition in human SMCs and in a C3H10T1/2 model, SMC differentiation was inhibited in both cases. Further characterization of downstream Notch signaling pathways showed activation of the c-Jun N-terminal kinase, p38 mitogen-activated protein kinase, and PI3K/Akt pathways. The activation of these pathways was sensitive to HDAC inhibition and was positively correlated with the differentiated phenotype. Conclusions Our studies define novel signaling pathways downstream of Notch signaling in human SMCs. In addition to the canonical CBF-1 pathway, Notch stimulates c-Jun N-terminal kinase, mitogen-activated protein kinase, and PI3K cascades. Both canonical and noncanonical pathways downstream of Notch promote a differentiated, contractile phenotype in SMCs. Although CBF-1–mediated Notch signaling is not suppressed by HDAC inhibition, HDAC activity is required for Notch differentiation signals through mitogen-activated protein kinase and PI3K pathways in SMCs. (J Am Heart Assoc. 2012;1:e000901 doi: 10.1161/JAHA.112.000901)
Collapse
Affiliation(s)
- Yuefeng Tang
- Center for Molecular Medicine, Maine Medical Center Research Institute Scarborough, ME
| | | | | |
Collapse
|
46
|
Michael DR, Salter RC, Ramji DP. TGF-β inhibits the uptake of modified low density lipoprotein by human macrophages through a Smad-dependent pathway: a dominant role for Smad-2. Biochim Biophys Acta Mol Basis Dis 2012; 1822:1608-16. [PMID: 22705205 PMCID: PMC3497875 DOI: 10.1016/j.bbadis.2012.06.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 05/29/2012] [Accepted: 06/05/2012] [Indexed: 11/17/2022]
Abstract
The anti-atherogenic cytokine, TGF-β, plays a key role during macrophage foam cell formation by modulating the expression of key genes involved in the control of cholesterol homeostasis. Unfortunately, the molecular mechanisms underlying these actions of TGF-β remain poorly understood. In this study we examine the effect of TGF-β on macrophage cholesterol homeostasis and delineate the role of Smads-2 and ‐3 during this process. Western blot analysis showed that TGF-β induces a rapid phosphorylation-dependent activation of Smad-2 and ‐3 in THP-1 and primary human monocyte-derived macrophages. Small interfering RNA-mediated knockdown of Smad-2/3 expression showed that the TGF-β-mediated regulation of key genes implicated in the uptake of modified low density lipoproteins and the efflux of cholesterol from foam cells was Smad-dependent. Additionally, through the use of virally delivered Smad-2 and/or Smad-3 short hairpin RNA, we demonstrate that TGF-β inhibits the uptake of modified LDL by macrophages through a Smad-dependent mechanism and that the TGF-β-mediated regulation of CD36, lipoprotein lipase and scavenger receptor-A gene expression was dependent on Smad-2. These studies reveal a crucial role for Smad signaling, particularly Smad-2, in the inhibition of foam cell formation by TGF-β through the regulation of expression of key genes involved in the control of macrophage cholesterol homeostasis.
Collapse
Affiliation(s)
- Daryn R Michael
- Cardiff School of Biosciences, Cardiff University, Cardiff, UK.
| | | | | |
Collapse
|
47
|
Increased smooth muscle contractility in mice deficient for neuropilin 2. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:548-59. [PMID: 22688055 DOI: 10.1016/j.ajpath.2012.04.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 03/30/2012] [Accepted: 04/05/2012] [Indexed: 12/28/2022]
Abstract
Neuropilins (NRPs) are transmembrane receptors that bind class 3 semaphorins and VEGF family members to regulate axon guidance and angiogenesis. Although expression of NRP1 by vascular smooth muscle cells (SMCs) has been reported, NRP function in smooth muscle (SM) in vivo is unexplored. Using Nrp2(+/LacZ) and Nrp2(+/gfp) transgenic mice, we observed robust and sustained expression of Nrp2 in the SM compartments of the bladder and gut, but no expression in vascular SM, skeletal muscle, or cardiac muscle. This expression pattern was recapitulated in vitro using primary human SM cell lines. Alterations in cell morphology after treatment of primary visceral SMCs with the NRP2 ligand semaphorin-3F (SEMA3F) were accompanied by inhibition of RhoA activity and myosin light chain phosphorylation, as well as decreased cytoskeletal stiffness. Ex vivo contractility testing of bladder muscle strips exposed to electrical stimulation or soluble agonists revealed enhanced tension generation of tissues from mice with constitutive or SM-specific knockout of Nrp2, compared with controls. Mice lacking Nrp2 also displayed increased bladder filling pressures, as assessed by cystometry in conscious mice. Together, these findings identify Nrp2 as a mediator of prorelaxant stimuli in SMCs and suggest a novel function for Nrp2 as a regulator of visceral SM contractility.
Collapse
|
48
|
Williams C, Xie AW, Emani S, Yamato M, Okano T, Emani SM, Wong JY. A Comparison of Human Smooth Muscle and Mesenchymal Stem Cells as Potential Cell Sources for Tissue-Engineered Vascular Patches. Tissue Eng Part A 2012; 18:986-98. [DOI: 10.1089/ten.tea.2011.0172] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Corin Williams
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Angela W. Xie
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Sirisha Emani
- Department of Cardiovascular Surgery, Children's Hospital Boston, Boston, Massachusetts
| | - Masayuki Yamato
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Japan
| | - Sitaram M. Emani
- Department of Cardiovascular Surgery, Children's Hospital Boston, Boston, Massachusetts
| | - Joyce Y. Wong
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| |
Collapse
|
49
|
Zhao L, Hantash BM. TGF-β1 regulates differentiation of bone marrow mesenchymal stem cells. VITAMINS AND HORMONES 2012; 87:127-41. [PMID: 22127241 DOI: 10.1016/b978-0-12-386015-6.00042-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Mesenchymal stromal/stem cells (MSCs) are a small population of stromal cells present in most adult connective tissues, such as bone marrow, fat tissue, and umbilical cord blood. MSCs are maintained in a relative state of quiescence in vivo but, in response to a variety of physiological and pathological stimuli, are capable of proliferating then differentiating into osteoblasts, chondrocytes, adipocytes, or other mesoderm-type lineages like smooth muscle cells (SMCs) and cardiomyocytes. Multiple signaling networks orchestrate MSCs differentiating into functional mesenchymal lineages. Among these, transforming growth factor-β1 (TGF-β1) has emerged as a key player. Hence, we summarize the effects of TGF-β1 on differentiation of MSCs toward different lineages. TGF-β1 can induce either chondrogenic or SMC differentiation of MSCs in vitro. However, it requires cell-cell and cell-matrix interactions, similar to development of these tissues in vivo. The effect of TGF-β1-regulated osteogenic differentiation of MSCs in vitro depends on the specific culture conditions involved. TGF-β1 inhibits adipogenic differentiation of MSCs in monolayer culture. Using this information, we may optimize the culture conditions to differentiate MSCs into desired lineages.
Collapse
Affiliation(s)
- Longmei Zhao
- Escape Therapeutics, Inc., San Jose, California, USA
| | | |
Collapse
|
50
|
Guo X, Chen SY. Transforming growth factor-β and smooth muscle differentiation. World J Biol Chem 2012; 3:41-52. [PMID: 22451850 PMCID: PMC3312200 DOI: 10.4331/wjbc.v3.i3.41] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 01/19/2012] [Accepted: 01/26/2012] [Indexed: 02/05/2023] Open
Abstract
Transforming growth factor (TGF)-β family members are multifunctional cytokines regulating diverse cellular functions such as growth, adhesion, migration, apoptosis, and differentiation. TGF-βs elicit their effects via specific type I and type II serine/threonine kinase receptors and intracellular Smad transcription factors. Knockout mouse models for the different components of the TGF-β signaling pathway have revealed their critical roles in smooth muscle cell (SMC) differentiation. Genetic studies in humans have linked mutations in these signaling components to specific cardiovascular disorders such as aorta aneurysm and congenital heart diseases due to SMC defects. In this review, the current understanding of TGF-β function in SMC differentiation is highlighted, and the role of TGF-β signaling in SMC-related diseases is discussed.
Collapse
Affiliation(s)
- Xia Guo
- Xia Guo, Shi-You Chen, Department of Physiology and Pharmacology, University of Georgia, Athens, GA 30602, United States
| | | |
Collapse
|