1
|
Li P, Xie W, Wei H, Yang F, Chen Y, Li Y. Transcriptome Analyses of Liver Sinusoidal Endothelial Cells Reveal a Consistent List of Candidate Genes Associated with Endothelial Dysfunction and the Fibrosis Progression. Curr Issues Mol Biol 2024; 46:7997-8014. [PMID: 39194690 DOI: 10.3390/cimb46080473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
Liver fibrosis is an important step in the transformation of chronic liver disease into cirrhosis and liver cancer, and structural changes and functional disorders of liver sinusoidal endothelial cells (LSECs) are early events in the occurrence of liver fibrosis. Therefore, it is necessary to identify the key regulatory genes of endothelial dysfunction in the process of liver fibrosis to provide a reference for the diagnosis and treatment of liver fibrosis. In this study, we identified 230 common differentially expressed genes (Co-DEGs) by analyzing transcriptomic data of primary LSECs from three different liver fibrosis mouse models (carbon tetrachloride; choline-deficient, l-amino acid-defined diet; and nonalcoholic steatohepatitis). Enrichment analysis revealed that the Co-DEGs were mainly involved in regulating the inflammatory response, immune response, angiogenesis, formation and degradation of the extracellular matrix, and mediating chemokine-related pathways. A Venn diagram analysis was used to identify 17 key genes related to the progression of liver cirrhosis. Regression analysis using the Lasso-Cox method identified genes related to prognosis among these key genes: SOX4, LGALS3, SERPINE2, CD52, and LPXN. In mouse models of liver fibrosis (bile duct ligation and carbon tetrachloride), all five key genes were upregulated in fibrotic livers. This study identified key regulatory genes for endothelial dysfunction in liver fibrosis, namely SOX4, LGALS3, SERPINE2, CD52, and LPXN, which will provide new targets for the development of therapeutic strategies targeting endothelial dysfunction in LSECs and liver fibrosis.
Collapse
Affiliation(s)
- Penghui Li
- Center for Health Research, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Wenjie Xie
- Center for Health Research, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Hongjin Wei
- Center for Health Research, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Fan Yang
- Center for Health Research, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yan Chen
- Center for Health Research, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Yinxiong Li
- Center for Health Research, Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangzhou 510530, China
| |
Collapse
|
2
|
Filla MS, Meyer KK, Faralli JA, Peters DM. Overexpression and Activation of αvβ3 Integrin Differentially Affects TGFβ2 Signaling in Human Trabecular Meshwork Cells. Cells 2021; 10:1923. [PMID: 34440692 PMCID: PMC8394542 DOI: 10.3390/cells10081923] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/14/2021] [Accepted: 07/21/2021] [Indexed: 12/13/2022] Open
Abstract
Studies from our laboratory have suggested that activation of αvβ3 integrin-mediated signaling could contribute to the fibrotic-like changes observed in primary open angle glaucoma (POAG) and glucocorticoid-induced glaucoma. To determine how αvβ3 integrin signaling could be involved in this process, RNA-Seq analysis was used to analyze the transcriptomes of immortalized trabecular meshwork (TM) cell lines overexpressing either a control vector or a wild type (WT) or a constitutively active (CA) αvβ3 integrin. Compared to control cells, hierarchical clustering, PANTHER pathway and protein-protein interaction (PPI) analysis of cells overexpressing WT-αvβ3 integrin or CA-αvβ3 integrin resulted in a significant differential expression of genes encoding for transcription factors, adhesion and cytoskeleton proteins, extracellular matrix (ECM) proteins, cytokines and GTPases. Cells overexpressing a CA-αvβ3 integrin also demonstrated an enrichment for genes encoding proteins found in TGFβ2, Wnt and cadherin signaling pathways all of which have been implicated in POAG pathogenesis. These changes were not observed in cells overexpressing WT-αvβ3 integrin. Our results suggest that activation of αvβ3 integrin signaling in TM cells could have significant impacts on TM function and POAG pathogenesis.
Collapse
Affiliation(s)
- Mark S. Filla
- Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI 53705, USA; (M.S.F.); (K.K.M.); (J.A.F.)
| | - Kristy K. Meyer
- Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI 53705, USA; (M.S.F.); (K.K.M.); (J.A.F.)
| | - Jennifer A. Faralli
- Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI 53705, USA; (M.S.F.); (K.K.M.); (J.A.F.)
| | - Donna M. Peters
- Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI 53705, USA; (M.S.F.); (K.K.M.); (J.A.F.)
- Ophthalmology & Visual Sciences, University of Wisconsin, Madison, WI 53705, USA
| |
Collapse
|
3
|
Li Y, Gao S, Han Y, Song L, Kong Y, Jiao Y, Huang S, Du J, Li Y. Variants of Focal Adhesion Scaffold Genes Cause Thoracic Aortic Aneurysm. Circ Res 2020; 128:8-23. [PMID: 33092471 DOI: 10.1161/circresaha.120.317361] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RATIONALE Thoracic aortic aneurysm (TAA) leads to substantial mortality worldwide. Familial and syndromic TAAs are highly correlated with genetics. However, the incidence of sporadic isolated TAA (iTAA) is much higher, and the genetic contribution is not yet clear. OBJECTIVE Here, we examined the genetic characteristics of sporadic iTAA. METHODS AND RESULTS We performed a genetic screen of 551 sporadic iTAA cases and 1071 controls via whole-exome sequencing. The prevalence of pathogenic mutations in known causal genes was 5.08% in the iTAA cohort. We selected 100 novel candidate genes using a strict strategy, and the suspected functional variants of these genes were significantly enriched in cases compared with controls and carried by 60.43% of patients. We found more severe phenotypes and a lower proportion of hypertension in cases with pathogenic mutations or suspected functional variants. Among the candidate genes, Testin (TES), which encodes a focal adhesion scaffold protein, was identified as a potential TAA causal gene, accounting for 4 patients with 2 missense variants in the LIM1 domain (c.751T>C encoding p.Y251H; c.838T>C encoding p.Y280H) and highly expressed in the aorta. The 2 variants led to a decrease in TES expression. The thoracic aorta was spontaneously dilated in the TesY249H knock-in and Tes-/- mice. Mechanistically, the p.Y249H variant or knockdown of TES led to the repression of vascular smooth muscle cell contraction genes and disturbed the vascular smooth muscle cell contractile phenotype. Interestingly, suspected functional variants of other focal adhesion scaffold genes, including TLN1 (Talin-1) and ZYX (zyxin), were also significantly enriched in patients with iTAA; moreover, their knockdown resulted in decreased contractility of vascular smooth muscle cells. CONCLUSIONS For the first time, this study revealed the genetic landscape across iTAA and showed that the focal adhesion scaffold genes are critical in the pathogenesis of iTAA.
Collapse
Affiliation(s)
- Yang Li
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Shijuan Gao
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Yingchun Han
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Li Song
- BGI Genomics, BGI-Shenzhen, China (Li Song)
| | - Yu Kong
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Yao Jiao
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Shan Huang
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Jie Du
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| | - Yulin Li
- Beijing Anzhen Hospital, Capital Medical University, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,Beijing Institute of Heart, Lung and Blood Vessel Disease, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li).,The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, China (Yang Li, S.G., Y.H., Y.K., Y.J., S.H., J.D., Yulin Li)
| |
Collapse
|
4
|
Bai X, Mangum K, Kakoki M, Smithies O, Mack CP, Taylor JM. GRAF3 serves as a blood volume-sensitive rheostat to control smooth muscle contractility and blood pressure. Small GTPases 2020; 11:194-203. [PMID: 29099324 PMCID: PMC7549679 DOI: 10.1080/21541248.2017.1375602] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Vascular resistance is a major determinant of BP and is controlled, in large part, by RhoA-dependent smooth muscle cell (SMC) contraction within small peripheral arterioles and previous studies from our lab indicate that GRAF3 is a critical regulator of RhoA in vascular SMC. The elevated contractile responses we observed in GRAF3 deficient vessels coupled with the hypertensive phenotype provided a mechanistic link for the hypertensive locus recently identified within the GRAF3 gene. On the basis of our previous findings that the RhoA signaling axis also controls SMC contractile gene expression and that GRAF3 expression was itself controlled by this pathway, we postulated that GRAF3 serves as an important counter-regulator of SMC phenotype. Indeed, our new findings presented herein indicate that GRAF3 expression acts as a pressure-sensitive rheostat to control vessel tone by both reducing calcium sensitivity and restraining expression of the SMC-specific contractile proteins that support this function. Collectively, these studies highlight the potential therapeutic value of GRAF3 in the control of human hypertension.
Collapse
Affiliation(s)
- Xue Bai
- Department of Pathology, University of North Carolina, Chapel Hill, NC, USA
| | - Kevin Mangum
- Department of Pathology, University of North Carolina, Chapel Hill, NC, USA
| | - Masao Kakoki
- Department of Pathology, University of North Carolina, Chapel Hill, NC, USA
| | - Oliver Smithies
- Department of Pathology, University of North Carolina, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Christopher P. Mack
- Department of Pathology, University of North Carolina, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Joan M. Taylor
- Department of Pathology, University of North Carolina, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
5
|
Zhu GH, Dai HP, Shen Q, Zhang Q. Downregulation of LPXN expression by siRNA decreases the malignant proliferation and transmembrane invasion of SHI-1 cells. Oncol Lett 2018; 17:135-140. [PMID: 30655748 PMCID: PMC6313184 DOI: 10.3892/ol.2018.9605] [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: 12/27/2017] [Accepted: 08/30/2018] [Indexed: 01/03/2023] Open
Abstract
The aim of the present study was to investigate the effects of decreasing leupaxin (LPXN) expression on the proliferation and invasion of human acute monocytic leukemia SHI-1 cells. The transfection efficiency of fluorescein amidite (FAM)-small interfering RNA (siRNA) was determined using flow cytometry, and the protein expression levels of LPXN, phosphorylated (p)-c-Jun N-terminal kinase (JNK), p-p38 mitogen-activated protein kinase (p38 MAPK) and p-extracellular-signal-regulated kinase (ERK) were detected by western blot analysis. Proliferation was determined using the cell counting kit-8 reagent and cellular transmembrane invasion ability was determined using a Transwell chamber system. The gelatinase levels of matrix metalloproteinase (MMP)-2 and MMP-9 in the cell culture supernatant were also analyzed by gelatin zymography. In SHI-1 cells, the optimal transfection conditions of siRNA were a cell density of 4×105 cells/ml and a ratio of siRNA/Lipofectamine® 2000 of 200 pmol/1 µl. The highest transfection efficiency of FAM-siRNA was 74.5%. In the present study, L2-siRNA was selected to effectively decrease the expression of LPXN. Following downregulation of LPXN expression by L2-siRNA, proliferation inhibition rates increased to 27.043±2.051 and cell transmembrane invasion rates decreased to 25.270±2.145 (P<0.05). The results of the western blot analysis and the gelatin zymography indicated that downregulation of LPXN expression increased the expression of p-p38 MAPK and p-JNK, and attenuated the secretion levels of MMP-2 and MMP-9. However, downregulation of LPXN expression had no effect on p-ERK expression in SHI-1 cells. The results of the present study indicated that downregulation of LPXN expression decreased the malignant proliferation and transmembrane invasion of SHI-1 cells by activating JNK and p38 MAPK, and inhibiting MMP-2 and MMP-9 secretion.
Collapse
Affiliation(s)
- Guo-Hua Zhu
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Hai-Ping Dai
- Leukemia Research Unit, Jiangsu Institute of Hematology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Qun Shen
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China.,Department of Hematology, First Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210009, P.R. China
| | - Qi Zhang
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| |
Collapse
|
6
|
朱 国, 戴 海, 段 元, 余 泽. [Small interfering RNA-mediated LPXN silencing suppresses proliferation and enhances drug sensitivity of human acute monocytic leukemia SHI-1 cells in vitro]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:807-811. [PMID: 33168498 PMCID: PMC6765540 DOI: 10.3969/j.issn.1673-4254.2018.07.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To investigate the effect of silencing LPXN expression by RNA interference on the proliferation and drug sensitivity of human acute monocytic leukemia SHI-1 cells in vitro. METHODS Small interfering RNA (siRNA) sequences targeting LPXN were designed and transiently transfected in SHI-1 cells via Lipofectamine 2000, and the most efficient siRNA sequence for LPXN silencing was identified using Western blotting. The protein expression levels of LPXN, p-JNK, p-P38 MAPK and p-ERK were in the cells transfected with the selected siRNA were detected using Western blotting, and the cell proliferation changes were assessed using CCK-8 reagent. RESULTS LPXN silencing by siRNA transfection resulted in significant proliferation suppression in SHI-1 cells with an inhibition rate of(27.04±2.05) % (P < 0.05). Western blotting showed that treatment of the siRNA-transfected SHI-1 cells with 0-25 μmol/L curcumin or with 0-2.0 μmol/L Ara-C further increased the cell inhibition rate and obviously enhanced the expressions of p-P38 MAPK and p-JNK without significantly affecting p-ERK expression. CONCLUSIONS Down-regulation of LPXN expression by siRNA transfection can suppress the proliferation and increase the drug sensitivity of SHI-1 cells probably by activating JNK and P38 MAPK.
Collapse
Affiliation(s)
- 国华 朱
- 南京中医药大学第一临床医学院,江苏 南京 210023First Clinical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - 海萍 戴
- 苏州大学第一附属医院血液科,江苏 苏州 215006Department of Hematology, First Hospital Affiliated to Suzhou University, Suzhou 215006, China
| | - 元勋 段
- 南京中医药大学第一临床医学院,江苏 南京 210023First Clinical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - 泽霖 余
- 南京中医药大学第一临床医学院,江苏 南京 210023First Clinical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
7
|
Oh RS, Haak AJ, Smith KMJ, Ligresti G, Choi KM, Xie T, Wang S, Walters PR, Thompson MA, Freeman MR, Manlove LJ, Chu VM, Feghali-Bostwick C, Roden AC, Schymeinsky J, Pabelick CM, Prakash YS, Vassallo R, Tschumperlin DJ. RNAi screening identifies a mechanosensitive ROCK-JAK2-STAT3 network central to myofibroblast activation. J Cell Sci 2018; 131:jcs.209932. [PMID: 29678906 DOI: 10.1242/jcs.209932] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 04/13/2018] [Indexed: 12/14/2022] Open
Abstract
Myofibroblasts play key roles in wound healing and pathological fibrosis. Here, we used an RNAi screen to characterize myofibroblast regulatory genes, using a high-content imaging approach to quantify α-smooth muscle actin stress fibers in cultured human fibroblasts. Screen hits were validated on physiological compliance hydrogels, and selected hits tested in primary fibroblasts from patients with idiopathic pulmonary fibrosis. Our RNAi screen led to the identification of STAT3 as an essential mediator of myofibroblast activation and function. Strikingly, we found that STAT3 phosphorylation, while responsive to exogenous ligands on both soft and stiff matrices, is innately active on a stiff matrix in a ligand/receptor-independent, but ROCK- and JAK2-dependent fashion. These results demonstrate how a cytokine-inducible signal can become persistently activated by pathological matrix stiffening. Consistent with a pivotal role for this pathway in driving persistent fibrosis, a STAT3 inhibitor attenuated murine pulmonary fibrosis when administered in a therapeutic fashion after bleomycin injury. Our results identify novel genes essential for the myofibroblast phenotype, and point to STAT3 as an important target in pulmonary fibrosis and other fibrotic diseases.
Collapse
Affiliation(s)
- Raymond S Oh
- Department of Environmental Health, Harvard School of Public Health, Boston, MA 02115, USA
| | - Andrew J Haak
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Karry M J Smith
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Giovanni Ligresti
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Kyoung Moo Choi
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Tiao Xie
- Image and Data Analysis Core, Harvard Medical School, Boston, MA 02115, USA
| | - Shaohua Wang
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Paula R Walters
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Michael A Thompson
- Departments of Anesthesiology and Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Michelle R Freeman
- Departments of Anesthesiology and Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Logan J Manlove
- Departments of Anesthesiology and Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Vivian M Chu
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Carol Feghali-Bostwick
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Anja C Roden
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jürgen Schymeinsky
- Department of Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach, Germany
| | - Christina M Pabelick
- Departments of Anesthesiology and Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Y S Prakash
- Departments of Anesthesiology and Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Robert Vassallo
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Daniel J Tschumperlin
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
8
|
Dierks S, von Hardenberg S, Schmidt T, Bremmer F, Burfeind P, Kaulfuß S. Leupaxin stimulates adhesion and migration of prostate cancer cells through modulation of the phosphorylation status of the actin-binding protein caldesmon. Oncotarget 2016; 6:13591-606. [PMID: 26079947 PMCID: PMC4537036 DOI: 10.18632/oncotarget.3792] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 03/18/2015] [Indexed: 12/15/2022] Open
Abstract
The focal adhesion protein leupaxin (LPXN) is overexpressed in a subset of prostate cancers (PCa) and is involved in the progression of PCa. In the present study, we analyzed the LPXN-mediated adhesive and cytoskeletal changes during PCa progression. We identified an interaction between the actin-binding protein caldesmon (CaD) and LPXN and this interaction is increased during PCa cell migration. Furthermore, knockdown of LPXN did not affect CaD expression but reduced CaD phosphorylation. This is known to destabilize the affinity of CaD to F-actin, leading to dynamic cell structures that enable cell motility. Thus, downregulation of CaD increased migration and invasion of PCa cells. To identify the kinase responsible for the LPXN-mediated phosphorylation of CaD, we used data from an antibody array, which showed decreased expression of TGF-beta-activated kinase 1 (TAK1) after LPXN knockdown in PC-3 PCa cells. Subsequent analyses of the downstream kinases revealed the extracellular signal-regulated kinase (ERK) as an interaction partner of LPXN that facilitates CaD phosphorylation during LPXN-mediated PCa cell migration. In conclusion, we demonstrate that LPXN directly influences cytoskeletal dynamics via interaction with the actin-binding protein CaD and regulates CaD phosphorylation by recruiting ERK to highly dynamic structures within PCa cells.
Collapse
Affiliation(s)
- Sascha Dierks
- Institute of Human Genetics, University Medical Center Göttingen, Germany
| | - Sandra von Hardenberg
- Institute of Human Genetics, University Medical Center Göttingen, Germany.,Center of Pharmacology and Toxicology, Hannover Medical School, Germany
| | - Thomas Schmidt
- Institute of Human Genetics, University Medical Center Göttingen, Germany.,Department of Anatomy, University of Witten/Herdecke, Witten, Germany
| | - Felix Bremmer
- Institute of Pathology, University Medical Center Göttingen, Germany
| | - Peter Burfeind
- Institute of Human Genetics, University Medical Center Göttingen, Germany
| | - Silke Kaulfuß
- Institute of Human Genetics, University Medical Center Göttingen, Germany
| |
Collapse
|
9
|
Kaulfuss S, Herr AM, Büchner A, Hemmerlein B, Günthert AR, Burfeind P. Leupaxin is expressed in mammary carcinoma and acts as a transcriptional activator of the estrogen receptor α. Int J Oncol 2015; 47:106-14. [PMID: 25955236 PMCID: PMC4485646 DOI: 10.3892/ijo.2015.2988] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 03/23/2015] [Indexed: 02/07/2023] Open
Abstract
Leupaxin belongs to the group of paxillin proteins and was reported to play a major role in the invasion and migration of prostate cancer cells. In the present study we were able to show by using a cDNA cancer profiling array that leupaxin is upregulated in breast and endometrial cancer, whereas downregulation of leupaxin was observed in lung cancer. In addition, immunohistochemical studies using a leupaxin-specific antibody on human breast cancer specimens (n=127) revealed that leupaxin is expressed mainly in invasive ductal carcinomas and ductal carcinoma in situ (40 and 49% respectively), and only in a minority of lobular mammary carcinomas. To further investigate the role of leupaxin in the progression of breast cancer the expression of leupaxin was analysed in six breast cancer cell lines. The estrogen receptor α (ERα)-positive HCC70 and the ERα-negative MDA-MB-231 cells showed leupaxin expression on the RNA and protein level. Leupaxin localizes in these mammary carcinoma cells at focal adhesion sites and shuttles between membrane and nucleus via its LD4 motif as major nuclear export signal. Interaction partners of leupaxin in the nucleus represent the estrogen receptors ERα and ERβ. Both ERα and ERβ bind to the LIM domains of leupaxin via their AF-1/DNA binding domains. Furthermore, leupaxin is able to induce transcriptional activity of ERα independent of the presence of estradiol. The specific downregulation of leupaxin expression using siRNAs in mammary carcinoma cells resulted in reduced migratory capability and diminished invasiveness whereas no effect on proliferation was observed. Collectively, these results show that leupaxin has particular influence on the progression and invasion of breast cancer cells and may therefore represent an interesting candidate protein for diagnosis and therapeutic interventions.
Collapse
Affiliation(s)
- Silke Kaulfuss
- Institute of Human Genetics, University Medical Center Göttingen, Germany
| | - Anna-Maria Herr
- Institute of Human Genetics, University Medical Center Göttingen, Germany
| | - Anja Büchner
- Institute of Human Genetics, University Medical Center Göttingen, Germany
| | | | - Andreas R Günthert
- Department of Gynaecology and Obstetrics, University Medical Center Göttingen, Germany
| | - Peter Burfeind
- Institute of Human Genetics, University Medical Center Göttingen, Germany
| |
Collapse
|
10
|
Veith C, Zakrzewicz D, Dahal BK, Bálint Z, Murmann K, Wygrecka M, Seeger W, Schermuly RT, Weissmann N, Kwapiszewska G. Hypoxia- or PDGF-BB-dependent paxillin tyrosine phosphorylation in pulmonary hypertension is reversed by HIF-1α depletion or imatinib treatment. Thromb Haemost 2014; 112:1288-303. [PMID: 25231004 DOI: 10.1160/th13-12-1031] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 07/10/2014] [Indexed: 12/27/2022]
Abstract
Chronic exposure to hypoxia induces a pronounced remodelling of the pulmonary vasculature leading to pulmonary hypertension (PH). The remodelling process also entails increased proliferation and decreased apoptosis of pulmonary arterial smooth muscle cells (PASMC), processes regulated by the cytoskeletal protein paxillin. In this study, we aimed to examine the molecular mechanisms leading to deregulation of paxillin in PH. We detected a time-dependent increase in paxillin tyrosine 31 (Y31) and 118 (Y118) phosphorylation following hypoxic exposure (1 % O2) or platelet-derived growth factor (PDGF)-BB stimulation of primary human PASMC. In addition, both, hypoxia- and PDGF-BB increased the nuclear localisation of phospho-paxillin Y31 as indicated by immunofluorescence staining in human PASMC. Elevated paxillin tyrosine phosphorylation in human PASMC was attenuated by hypoxia-inducible factor (HIF)-1α depletion or by treatment with the PDGF-BB receptor antagonist, imatinib. Moreover, we observed elevated paxillin Y31 and Y118 phosphorylation in the pulmonary vasculature of chronic hypoxic mice (21 days, 10 % O2) which was reversible by imatinib-treatment. PDGF-BB-dependent PASMC proliferation was regulated via the paxillin-Erk1/2-cyclin D1 pathway. In conclusion, we suggest paxillin up-regulation and phosphorylation as an important mechanism of vascular remodelling underlying pulmonary hypertension.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - N Weissmann
- Norbert Weissmann, Excellence Cluster Cardio-Pulmonary System (ECCPS), Justus-Liebig-University Giessen, Aulweg 130, 35392 Giessen, Germany, Tel.: +49 641 99 46000, Fax: +49 641 99 42419, E-mail:
| | | |
Collapse
|
11
|
Bai X, Lenhart KC, Bird KE, Suen AA, Rojas M, Kakoki M, Li F, Smithies O, Mack CP, Taylor JM. The smooth muscle-selective RhoGAP GRAF3 is a critical regulator of vascular tone and hypertension. Nat Commun 2013; 4:2910. [PMID: 24335996 PMCID: PMC4237314 DOI: 10.1038/ncomms3910] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Accepted: 11/11/2013] [Indexed: 12/13/2022] Open
Abstract
Although hypertension is a worldwide health issue, an incomplete understanding of its aetiology has hindered our ability to treat this complex disease. Here we identify arhgap42 (also known as GRAF3) as a Rho-specific GAP expressed specifically in smooth muscle cells (SMCs) in mice and humans. We show that GRAF3-deficient mice exhibit significant hypertension and increased pressor responses to angiotensin II and endothelin-1; these effects are prevented by treatment with the Rho-kinase inhibitor, Y27632. RhoA activity and myosin light chain phosphorylation are elevated in GRAF3-depleted SMCs in vitro and in vivo, and isolated vessel segments from GRAF3-deficient mice show increased contractility. Taken together, our data indicate that GRAF3-mediated inhibition of RhoA activity in vascular SMCs is necessary for maintaining normal blood pressure homoeostasis. Moreover, these findings provide a potential mechanism for a hypertensive locus recently identified within arhgap42 and provide a foundation for the future development of innovative hypertension therapies.
Collapse
Affiliation(s)
- Xue Bai
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Kaitlin C. Lenhart
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Kim E. Bird
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Alisa A. Suen
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Mauricio Rojas
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Masao Kakoki
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Feng Li
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Oliver Smithies
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Christopher P. Mack
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Joan M. Taylor
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
12
|
Deakin NO, Pignatelli J, Turner CE. Diverse roles for the paxillin family of proteins in cancer. Genes Cancer 2012; 3:362-70. [PMID: 23226574 DOI: 10.1177/1947601912458582] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The paxillin family of intracellular scaffold proteins includes paxillin, Hic-5, and leupaxin, and all have been identified as key regulators of the cellular migration machinery in both 2- and 3-dimensional microenvironments. Herein, we provide insight into the roles of these proteins during tumorigenesis and metastasis, highlighting their functions in cancer initiation as well as tumor cell dissemination and survival. Furthermore, we speculate on the potential of paxillin family proteins as both future prognostic and therapeutic targets.
Collapse
Affiliation(s)
- Nicholas O Deakin
- State University of New York Upstate Medical University, Syracuse, NY, USA
| | | | | |
Collapse
|
13
|
Xu Z, Ji G, Shen J, Wang X, Zhou J, Li L. SOX9 and myocardin counteract each other in regulating vascular smooth muscle cell differentiation. Biochem Biophys Res Commun 2012; 422:285-90. [PMID: 22580282 DOI: 10.1016/j.bbrc.2012.04.149] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 04/26/2012] [Indexed: 02/06/2023]
Abstract
Transdifferentiation of vascular smooth muscle cells (VSMC) into chondrogenic cells contributes significantly to vascular calcification during the pathogenesis of atherosclerosis. However, the transcriptional mechanisms that control such phenotypic switch remain unclear. This process is characterized by the induction of Sox9 and Col2a1 genes accompanied by the repression of myocardin (Myocd) and SMC differentiation markers such as SM22, SM α-actin and SM-MHC. Here we explore the regulatory role of SOX9, the master regulator for chondrogenesis, in modulating SMC marker gene expression. qRT-PCR and luciferase assays show that over-expression of SOX9 inhibits SMC gene transcription and promoter activities induced by myocardin, the master regulator of smooth muscle differentiation. Such suppression is independent of the CArG box in the SMC promoters but dependent on myocardin. EMSA assay further shows that SOX9 neither participates in SRF (serum response factor) binding to the CArG box nor interacts with SRF, while co-immunoprecipitation demonstrates an association of SOX9 with myocardin. Conversely, myocardin suppresses SOX9-mediated chondrogenic gene Col2a1 expression. These findings provide the first mechanistic insights into the important regulatory role of SOX9 and myocardin in controlling the transcription program during SMC transdifferentiation into chondrocytes.
Collapse
Affiliation(s)
- Zhonghui Xu
- Department of Internal Medicine, Wayne State University, Detroit, MI 48201, USA
| | | | | | | | | | | |
Collapse
|
14
|
|
15
|
Mack CP. Signaling mechanisms that regulate smooth muscle cell differentiation. Arterioscler Thromb Vasc Biol 2011; 31:1495-505. [PMID: 21677292 PMCID: PMC3141215 DOI: 10.1161/atvbaha.110.221135] [Citation(s) in RCA: 194] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 04/25/2011] [Indexed: 01/05/2023]
Abstract
Extensive studies over the last 30 years have demonstrated that vascular smooth muscle cell (SMC) differentiation and phenotypic modulation is controlled by a dynamic array of environmental cues. The identification of the signaling mechanisms by which these environmental cues regulate SMC phenotype has been more difficult because of our incomplete knowledge of the transcription mechanisms that regulate SMC-specific gene expression. However, recent advances in this area have provided significant insight, and the goal of this review is to summarize the signaling mechanisms by which extrinsic cues control SMC differentiation.
Collapse
Affiliation(s)
- Christopher P Mack
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599-7525, USA.
| |
Collapse
|
16
|
Majesky MW, Dong XR, Regan JN, Hoglund VJ. Vascular smooth muscle progenitor cells: building and repairing blood vessels. Circ Res 2011; 108:365-77. [PMID: 21293008 PMCID: PMC3382110 DOI: 10.1161/circresaha.110.223800] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 12/27/2010] [Indexed: 01/17/2023]
Abstract
Molecular pathways that control the specification, migration, and number of available smooth muscle progenitor cells play key roles in determining blood vessel size and structure, capacity for tissue repair, and progression of age-related disorders. Defects in these pathways produce malformations of developing blood vessels, depletion of smooth muscle progenitor cell pools for vessel wall maintenance and repair, and aberrant activation of alternative differentiation pathways in vascular disease. A better understanding of the molecular mechanisms that uniquely specify and maintain vascular smooth muscle cell precursors is essential if we are to use advances in stem and progenitor cell biology and somatic cell reprogramming for applications directed to the vessel wall.
Collapse
Affiliation(s)
- Mark W Majesky
- Seattle Children's Research Institute, University of Washington, 1900 Ninth Ave, M/S C9S-5, Seattle, WA 98101, USA.
| | | | | | | |
Collapse
|
17
|
Tanaka T, Moriwaki K, Murata S, Miyasaka M. LIM domain-containing adaptor, leupaxin, localizes in focal adhesion and suppresses the integrin-induced tyrosine phosphorylation of paxillin. Cancer Sci 2010; 101:363-8. [PMID: 19917054 PMCID: PMC11158308 DOI: 10.1111/j.1349-7006.2009.01398.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Focal adhesion (FA) consists of multiple cellular proteins including paxillin and serves as a center for adhesion-mediated signaling. The assembly and disassembly of FAs is regulated by locally produced intracellular signals, and tyrosine phosphorylation of paxillin has been implicated in this process. A Lin-11 Isl-1 Mec-3 (LIM) domain-containing adaptor protein, leupaxin, a member of the paxillin family, is expressed in leukocytes as well as in certain cancer cells, and shares overall structural characteristics with paxillin. However, it remains unknown whether leupaxin and paxillin cooperate with or antagonize each other in integrin signaling. Here we show that leupaxin potently represses the tyrosine phosphorylation of paxillin. When expressed in mouse thymoma BW5147 cells bound to ICAM-1, leupaxin accumulated in FA-like patches in the cell periphery. When expressed in NIH3T3 and HEK293T cells, leupaxin localized to FAs upon cell adhesion to fibronectin and strongly suppressed the integrin-induced tyrosine phosphorylation of paxillin. In integrin-stimulated HEK293T cells, leupaxin's LIM3 domain appeared essential for selective FA localization and the suppression of paxillin tyrosine phosphorylation. Leupaxin's LD3 motif, which is critical for stable association with FAK, was dispensable for leupaxin's suppressive ability. In addition, leupaxin reduced the spreading of NIH3T3 cells on fibronectin, which required both the LD3 motif and LIM3 domain. When expressed in human leukocytic K562 cells, leupaxin significantly suppressed integrin alpha5beta1-mediated cell adhesion to fibronectin and the tyrosine phosphorylation of paxillin. These findings indicate that leupaxin functions as a paxillin counterpart that potently suppresses the tyrosine phosphorylation of paxillin during integrin signaling.
Collapse
Affiliation(s)
- Toshiyuki Tanaka
- Laboratory of Immunodynamics, Department of Microbiology and Immunology, Osaka University, Graduate School of Medicine, Osaka, Japan.
| | | | | | | |
Collapse
|
18
|
Neuman NA, Ma S, Schnitzler GR, Zhu Y, Lagna G, Hata A. The four-and-a-half LIM domain protein 2 regulates vascular smooth muscle phenotype and vascular tone. J Biol Chem 2009; 284:13202-12. [PMID: 19265191 PMCID: PMC2676052 DOI: 10.1074/jbc.m900282200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Revised: 02/23/2009] [Indexed: 01/14/2023] Open
Abstract
In response to vascular injury, differentiated vascular smooth muscle cells (vSMCs) undergo a unique process known as "phenotype modulation," transitioning from a quiescent, "contractile" phenotype to a proliferative, "synthetic" state. We have demonstrated previously that the signaling pathway of bone morphogenetic proteins, members of the transforming growth factor beta family, play a role in the induction and maintenance of a contractile phenotype in human primary pulmonary artery smooth muscle cells. In this study, we show that a four-and-a-half LIM domain protein 2 (FHL2) inhibits transcriptional activation of vSMC-specific genes mediated by the bone morphogenetic protein signaling pathway through the CArG box-binding proteins, such as serum response factor and members of the myocardin (Myocd) family. Interestingly, FHL2 does not affect recruitment of serum response factor or Myocd, however, it inhibits recruitment of a component of the SWI/SNF chromatin remodeling complex, Brg1, and RNA polymerase II, which are essential for the transcriptional activation. This is a novel mechanism of regulation of SMC-specific contractile genes by FHL2. Finally, aortic rings from homozygous FHL2-null mice display abnormalities in both endothelial-dependent and -independent relaxation, suggesting that FHL2 is essential for the regulation of vasomotor tone.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Aorta, Thoracic/cytology
- Aorta, Thoracic/metabolism
- Blotting, Western
- Bone Morphogenetic Protein Receptors, Type II/genetics
- Bone Morphogenetic Protein Receptors, Type II/metabolism
- COS Cells
- Cells, Cultured
- Chlorocebus aethiops
- Chromatin Assembly and Disassembly
- Chromatin Immunoprecipitation
- DNA Helicases/genetics
- DNA Helicases/metabolism
- Fluorescent Antibody Technique
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Homeodomain Proteins/physiology
- Humans
- LIM-Homeodomain Proteins
- Mice
- Mice, Knockout
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Muscle Proteins/physiology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Phenotype
- Promoter Regions, Genetic
- Pulmonary Artery/cytology
- Pulmonary Artery/metabolism
- RNA Polymerase II/genetics
- RNA Polymerase II/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Serum Response Factor/genetics
- Serum Response Factor/metabolism
- Signal Transduction
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription Factors/physiology
- Transcriptional Activation
Collapse
Affiliation(s)
- Nicole A Neuman
- Department of Biochemistry, Tufts University School of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | | | | | | | | | | |
Collapse
|