1
|
Wang G, Zhao P, Yin C, Zheng X, Xie Y, Li X, Shang D, Shao S, Chen H, Wei L, Song Z. KIF11 promotes vascular smooth muscle cell proliferation by regulating cell cycle progression and accelerates neointimal formation after arterial injury in mice. Front Pharmacol 2024; 15:1392352. [PMID: 39166113 PMCID: PMC11333341 DOI: 10.3389/fphar.2024.1392352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
Background and aims: One of the primary causes of lumen narrowing is vascular injury induced during medical procedures. Vascular injury disrupts the integrity of the endothelium, triggering platelet deposition, leukocyte recruitment, and the release of inflammatory factors. This, in turn, induces the proliferation of vascular smooth muscle cells (VSMCs), leading to neointima formation. However, the molecular mechanism underlying VSMC proliferation following injury remains unknown. KIF11 is critical in regulating the cell cycle by forming bipolar spindles during mitotic metaphase. This process may contribute to VSMCs proliferation and neointima formation following vascular injury. Yet, the function of KIF11 in VSMCs has not been elucidated. This study aims to investigate the role and mechanisms of KIF11 in regulating VSMCs cycle progression and proliferation. Methods: After conducting biological analysis of the transcriptome sequencing data from the mouse carotid artery injury model and the cell transcriptome data of PDGF-BB-induced VSMCs, we identified a potential target gene, KIF11, which may play a crucial role in vascular injury. Then we established a vascular injury model to investigate how changes in KIF11 expression and activity influence in vivo VSMCs proliferation and neointimal formation. In addition, we employed siRNA and specific inhibitors to suppress KIF11 expression and activity in VSMCs cultured in vitro to study the mechanisms underlying VSMCs cycle progression and proliferation. Results: The results of immunohistochemistry and immunofluorescence indicate a significant upregulation of KIF11 expression in the injured vascular. The intraperitoneal injection of the KIF11 specific inhibitor, K858, partially inhibits intimal hyperplasia in the vascular injury model. In vitro experiments further demonstrate that PDGF-BB upregulates KIF11 expression through the PI3K/AKT pathway, and enhances KIF11 activity. Inhibition of both KIF11 expression and activity partially reverses the pro-cycle progression and pro-proliferation effects of PDGF-BB on VSMCs. Additionally, KIF11 overexpression partially counteracts the proliferation arrest and cell cycle arrest induced by inhibiting the PI3K/AKT pathway in VSMCs. Conclusion: Our study highlights the crucial role of KIF11 in regulating the cycle progression and proliferation of VSMCs after vascular injury. A comprehensive understanding of these mechanisms could pave the way for potential therapeutic interventions in treating vascular stenosis.
Collapse
Affiliation(s)
- Gengqiao Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Peng Zhao
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chuanzheng Yin
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xichuan Zheng
- Department of Vascular Surgery, The Southwest Hospital of AMU, Chongqing, China
| | - Yuhang Xie
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuan Li
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dan Shang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuyu Shao
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hao Chen
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Li Wei
- Department of Gerontology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zifang Song
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
2
|
Iram T, Garcia MA, Amand J, Kaur A, Atkins M, Iyer M, Lam M, Ambiel N, Jorgens DM, Keller A, Wyss-Coray T, Kern F, Zuchero JB. SRF transcriptionally regulates the oligodendrocyte cytoskeleton during CNS myelination. Proc Natl Acad Sci U S A 2024; 121:e2307250121. [PMID: 38483990 PMCID: PMC10962977 DOI: 10.1073/pnas.2307250121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 02/10/2024] [Indexed: 03/19/2024] Open
Abstract
Myelination of neuronal axons is essential for nervous system development. Myelination requires dramatic cytoskeletal dynamics in oligodendrocytes, but how actin is regulated during myelination is poorly understood. We recently identified serum response factor (SRF)-a transcription factor known to regulate expression of actin and actin regulators in other cell types-as a critical driver of myelination in the aged brain. Yet, a major gap remains in understanding the mechanistic role of SRF in oligodendrocyte lineage cells. Here, we show that SRF is required cell autonomously in oligodendrocytes for myelination during development. Combining ChIP-seq with RNA-seq identifies SRF-target genes in oligodendrocyte precursor cells and oligodendrocytes that include actin and other key cytoskeletal genes. Accordingly, SRF knockout oligodendrocytes exhibit dramatically reduced actin filament levels early in differentiation, consistent with its role in actin-dependent myelin sheath initiation. Surprisingly, oligodendrocyte-restricted loss of SRF results in upregulation of gene signatures associated with aging and neurodegenerative diseases. Together, our findings identify SRF as a transcriptional regulator that controls the expression of cytoskeletal genes required in oligodendrocytes for myelination. This study identifies an essential pathway regulating oligodendrocyte biology with high relevance to brain development, aging, and disease.
Collapse
Affiliation(s)
- Tal Iram
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA94305
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Miguel A. Garcia
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA94305
| | - Jérémy Amand
- Department of Clinical Bioinformatics, Helmholtz Institute for Pharmaceutical Research Saarland–Helmholtz Centre for Infection Research, Saarland University Campus, Saarbrücken66123, Germany
- Clinical Bioinformatics, Saarland University, Saarbrücken66123, Germany
| | - Achint Kaur
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA94305
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Micaiah Atkins
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA94305
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Manasi Iyer
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA94305
| | - Mable Lam
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA94305
| | - Nicholas Ambiel
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA94305
| | | | - Andreas Keller
- Department of Clinical Bioinformatics, Helmholtz Institute for Pharmaceutical Research Saarland–Helmholtz Centre for Infection Research, Saarland University Campus, Saarbrücken66123, Germany
- Clinical Bioinformatics, Saarland University, Saarbrücken66123, Germany
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA94305
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Fabian Kern
- Department of Clinical Bioinformatics, Helmholtz Institute for Pharmaceutical Research Saarland–Helmholtz Centre for Infection Research, Saarland University Campus, Saarbrücken66123, Germany
- Clinical Bioinformatics, Saarland University, Saarbrücken66123, Germany
| | - J. Bradley Zuchero
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
3
|
Zhang W, Wu Y, J Gunst S. Membrane adhesion junctions regulate airway smooth muscle phenotype and function. Physiol Rev 2023; 103:2321-2347. [PMID: 36796098 PMCID: PMC10243546 DOI: 10.1152/physrev.00020.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 02/09/2023] [Accepted: 02/15/2023] [Indexed: 02/18/2023] Open
Abstract
The local environment surrounding airway smooth muscle (ASM) cells has profound effects on the physiological and phenotypic properties of ASM tissues. ASM is continually subjected to the mechanical forces generated during breathing and to the constituents of its surrounding extracellular milieu. The smooth muscle cells within the airways continually modulate their properties to adapt to these changing environmental influences. Smooth muscle cells connect to the extracellular cell matrix (ECM) at membrane adhesion junctions that provide mechanical coupling between smooth muscle cells within the tissue. Membrane adhesion junctions also sense local environmental signals and transduce them to cytoplasmic and nuclear signaling pathways in the ASM cell. Adhesion junctions are composed of clusters of transmembrane integrin proteins that bind to ECM proteins outside the cell and to large multiprotein complexes in the submembranous cytoplasm. Physiological conditions and stimuli from the surrounding ECM are sensed by integrin proteins and transduced by submembranous adhesion complexes to signaling pathways to the cytoskeleton and nucleus. The transmission of information between the local environment of the cells and intracellular processes enables ASM cells to rapidly adapt their physiological properties to modulating influences in their extracellular environment: mechanical and physical forces that impinge on the cell, ECM constituents, local mediators, and metabolites. The structure and molecular organization of adhesion junction complexes and the actin cytoskeleton are dynamic and constantly changing in response to environmental influences. The ability of ASM to rapidly accommodate to the ever-changing conditions and fluctuating physical forces within its local environment is essential for its normal physiological function.
Collapse
Affiliation(s)
- Wenwu Zhang
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Yidi Wu
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Susan J Gunst
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| |
Collapse
|
4
|
PI3K Isoforms in Vascular Biology, A Focus on the Vascular System-Immune Response Connection. Curr Top Microbiol Immunol 2022; 436:289-309. [DOI: 10.1007/978-3-031-06566-8_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
5
|
Wu Y, Huang Y, Zhang W, Gunst SJ. The proprotein convertase furin inhibits IL-13-induced inflammation in airway smooth muscle by regulating integrin-associated signaling complexes. Am J Physiol Lung Cell Mol Physiol 2021; 321:L102-L115. [PMID: 34009050 DOI: 10.1152/ajplung.00618.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Furin is a proprotein convertase that regulates the activation and the inactivation of multiple proteins including matrix metalloproteinases, integrins, and cytokines. It is a serine endoprotease that localizes to the plasma membrane and can be secreted into the extracellular space. The role of furin in regulating inflammation in isolated canine airway smooth muscle tissues was investigated. The treatment of airway tissues with recombinant furin (rFurin) inhibited the activation of Akt and eotaxin secretion induced by IL-13, and it prevented the IL-13-induced suppression of smooth muscle myosin heavy chain expression. rFurin promoted a differentiated phenotype by activating β1-integrin proteins and stimulating the activation of the adhesome proteins vinculin and paxillin by talin. Activated paxillin induced the binding of Akt to β-parvin IPP [integrin-linked kinase (ILK), PINCH, parvin] complexes, which inhibits Akt activation. Treatment of tissues with a furin inhibitor or the depletion of endogenous furin using shRNA resulted in Akt activation and inflammatory responses similar to those induced by IL-13. Furin inactivation or IL-13 caused talin cleavage and integrin inactivation, resulting in the inactivation of vinculin and paxillin. Paxillin inactivation resulted in the coupling of Akt to α-parvin IPP complexes, which catalyze Akt activation and an inflammatory response. The results demonstrate that furin inhibits inflammation in airway smooth muscle induced by IL-13 and that the anti-inflammatory effects of furin are mediated by activating integrin proteins and integrin-associated signaling complexes that regulate Akt-mediated pathways to the nucleus. Furin may have therapeutic potential for the treatment of inflammatory conditions of the lungs and airways.
Collapse
Affiliation(s)
- Yidi Wu
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Youliang Huang
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Wenwu Zhang
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Susan J Gunst
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
6
|
Huang Y, Gunst SJ. Phenotype transitions induced by mechanical stimuli in airway smooth muscle are regulated by differential interactions of parvin isoforms with paxillin and Akt. Am J Physiol Lung Cell Mol Physiol 2020; 318:L1036-L1055. [PMID: 32130030 DOI: 10.1152/ajplung.00506.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Mechanical tension and humoral stimuli can induce transitions in airway smooth muscle phenotype between a synthetic inflammatory state that promotes cytokine secretion and a differentiated state that promotes the expression of smooth muscle phenotype-specific proteins. When tissues are maintained under high tension, Akt activation and eotaxin secretion are suppressed, but expression of the differentiation marker protein, smooth muscle myosin heavy chain (SmMHC), is promoted. When tissues are maintained under low tension, Akt activation and eotaxin secretion are stimulated, and the differentiated phenotype is suppressed. We hypothesized that mechanical stimuli are differentially transduced to Akt-mediated signaling pathways that regulate phenotype expression by α-parvin and β-parvin integrin-linked kinase/PINCH/parvin (IPP) signaling complexes within integrin adhesomes. High tension or ACh triggered paxillin phosphorylation and the binding of phospho-paxillin to β-parvin IPP complexes. This inhibited Akt activation and promoted SmMHC expression. Low tension or IL-4 did not elicit paxillin phosphorylation and triggered the binding of unphosphorylated paxillin to α-parvin IPP complexes, which promoted Akt activation and eotaxin secretion and suppressed SmMHC expression. Expression of a nonphosphorylatable paxillin mutant or β-parvin depletion by siRNA promoted the inflammatory phenotype, whereas the depletion of α-parvin promoted the differentiated phenotype. Results demonstrate that phenotype expression is regulated by the differential interaction of phosphorylated and unphosphorylated paxillin with α-parvin and β-parvin IPP complexes and that these complexes have opposite effects on the activation of Akt. Our results describe a novel molecular mechanism for transduction of mechanical and humoral stimuli within integrin signaling complexes to regulate phenotype expression in airway smooth muscle.
Collapse
Affiliation(s)
- Youliang Huang
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Susan J Gunst
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
7
|
Jhun H, Baek S, Kim J, Lee KP, Park HY, Park WH, Lim K, Kim J. Effect of Korean Magnolia obovata Extract on Platelet-Derived Growth Factor-Induced Vascular Smooth Muscle Cells. Chin J Integr Med 2019; 26:677-682. [PMID: 31571123 DOI: 10.1007/s11655-019-3171-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2018] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To investigate the effects of Korean Magnolia obovata crude extract (KME) on plateletderived growth factor (PDGF)-BB-induced proliferation and migration of vascular smooth muscle cells (VSMCs). METHODS KME composition was analyzed by high-performance liquid chromatography (HPLC). VSMCs were isolated from the aorta of a Sprague-Dawley rat, incubated in serum free-Dulbecco's modified Eagle's medium in the presence or absence of KME (10, 30, 100, and 300 μg/mL), then further treated with PDGF-BB (10 ng/mL). VSMC proliferation was detected using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and VSMC migration was determined using the Boyden chamber and scratch wound healing assays. Western blot analysis was used to detect phosphorylation of extracellular signal-regulated protein kinases 1 and 2 (p-ERK1/2), protein kinase B (p-Akt), and stress-activated protein kinase/c-Jun NH2-terminal kinase (p-SAPK/JNK). The antimigration and proliferation effects of KME were tested using aortic sprout outgrowth. RESULTS The HPLC analysis identified honokiol (0.45 mg/g) and magnolol (0.34 mg/g) as the major components of KME. KME (30, 100, and 300 μg/mL) significantly decreased the proliferation and migration of PDGF-BB-stimulated (10 ng/mL) VSMCs and the PDGF-BB-induced phosphorylation of EKR1/2, Akt, and SAPK/JNK (P<0.05). Furthermore, PDGF-BBinduced VSMCs treated with 300 μg/mL of KME showed reduction in aortic sprout outgrowth. CONCLUSION KME could inhibit abnormal proliferation and migration of VSMCs by down-regulating the phosphorylation of EKR1/2 and Akt. Thus, KME might be a functional food for preventing vascular disorders.
Collapse
Affiliation(s)
- Hyunjhung Jhun
- Technical Assistance Center, Korea Food Research Institute, Jeonbuk, 55365, Republic of Korea
| | - Suji Baek
- Department of Medical Science, School of Medicine, Konkuk University, Seoul, Republic of Korea
| | - Jinwoo Kim
- Department of Bio-Science, College of Natural Science, Dongguk University, Gyeongbuk, 38066, Republic of Korea
| | - Kang-Pa Lee
- Department of Bio-Science, College of Natural Science, Dongguk University, Gyeongbuk, 38066, Republic of Korea
| | - Hun-Young Park
- Physical Activity & Performance Institute, Konkuk University, Seoul, 05029, Republic of Korea
| | - Won-Hwan Park
- Department of Diagnostic, College of Korean Medicine, Dongguk University Goyang, Gyeonggi-do, 10326, Republic of Korea
| | - Kiwon Lim
- Physical Activity & Performance Institute, Konkuk University, Seoul, 05029, Republic of Korea
| | - Jisu Kim
- Physical Activity & Performance Institute, Konkuk University, Seoul, 05029, Republic of Korea.
| |
Collapse
|
8
|
Gibson ES, Woolfrey KM, Li H, Hogan PG, Nemenoff RA, Heasley LE, Dell'Acqua ML. Subcellular Localization and Activity of the Mitogen-Activated Protein Kinase Kinase 7 (MKK7) γ Isoform are Regulated through Binding to the Phosphatase Calcineurin. Mol Pharmacol 2018; 95:20-32. [PMID: 30404891 DOI: 10.1124/mol.118.113159] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 10/31/2018] [Indexed: 11/22/2022] Open
Abstract
Calcineurin (CaN) phosphatase signaling is regulated by targeting CaN to substrates, inhibitors, and scaffold proteins containing docking motifs with the consensus sequence of PxIxIT. Here, we identify the docking of CaN to the γ isoform of MKK7, a component of the c-Jun N-terminal kinase (JNK) pathway. Because of alternative splicing of a single exon within the N-terminal domain, MKK7γ encodes a unique PxIxIT motif (PIIVIT) that is not present in MKK7α or β We found that MKK7γ bound directly to CaN through this PIIVIT motif in vitro, immunoprecipitated with CaN from cell extracts, and exhibited fluorescence resonance energy transfer (FRET) with CaN in the cytoplasm but not in the nucleus of living cells. In contrast, MKK7α and β exhibited no direct binding or FRET with CaN and were localized more in the nucleus than the cytoplasm. Furthermore, the inhibition of CaN phosphatase activity increased the basal phosphorylation of MKK7γ but not MKK7β Deletion of the MKK7γ PIIVIT motif eliminated FRET with CaN and promoted MKK7γ redistribution to the nucleus; however, the inhibition of CaN activity did not alter MKK7γ localization, indicating that MKK7γ cytoplasmic retention by CaN is phosphatase activity independent. Finally, the inhibition of CaN phosphatase activity in vascular smooth muscle cells, which express MKK7γ mRNA, enhances JNK activation. Overall, we conclude that the MKK7γ-specific PxIxIT motif promotes high-affinity CaN binding that could promote novel cross talk between CaN and JNK signaling by limiting MKK7γ phosphorylation and restricting its localization to the cytoplasm.
Collapse
Affiliation(s)
- Emily S Gibson
- Department of Pharmacology (E.S.G., K.M.W., M.L.D.) and Department of Medicine, Division of Renal Diseases and Hypertension (R.A.N.), University of Colorado School of Medicine, Aurora, Colorado; Department of Craniofacial Biology, University of Colorado School of Dental Medicine, Aurora, Colorado (L.E.H.); Immune Disease Institute, Harvard Medical School, Boston, Massachusetts (H.L.); and La Jolla Institute for Allergy and Immunology, La Jolla, California (P.G.H.)
| | - Kevin M Woolfrey
- Department of Pharmacology (E.S.G., K.M.W., M.L.D.) and Department of Medicine, Division of Renal Diseases and Hypertension (R.A.N.), University of Colorado School of Medicine, Aurora, Colorado; Department of Craniofacial Biology, University of Colorado School of Dental Medicine, Aurora, Colorado (L.E.H.); Immune Disease Institute, Harvard Medical School, Boston, Massachusetts (H.L.); and La Jolla Institute for Allergy and Immunology, La Jolla, California (P.G.H.)
| | - Huiming Li
- Department of Pharmacology (E.S.G., K.M.W., M.L.D.) and Department of Medicine, Division of Renal Diseases and Hypertension (R.A.N.), University of Colorado School of Medicine, Aurora, Colorado; Department of Craniofacial Biology, University of Colorado School of Dental Medicine, Aurora, Colorado (L.E.H.); Immune Disease Institute, Harvard Medical School, Boston, Massachusetts (H.L.); and La Jolla Institute for Allergy and Immunology, La Jolla, California (P.G.H.)
| | - Patrick G Hogan
- Department of Pharmacology (E.S.G., K.M.W., M.L.D.) and Department of Medicine, Division of Renal Diseases and Hypertension (R.A.N.), University of Colorado School of Medicine, Aurora, Colorado; Department of Craniofacial Biology, University of Colorado School of Dental Medicine, Aurora, Colorado (L.E.H.); Immune Disease Institute, Harvard Medical School, Boston, Massachusetts (H.L.); and La Jolla Institute for Allergy and Immunology, La Jolla, California (P.G.H.)
| | - Raphael A Nemenoff
- Department of Pharmacology (E.S.G., K.M.W., M.L.D.) and Department of Medicine, Division of Renal Diseases and Hypertension (R.A.N.), University of Colorado School of Medicine, Aurora, Colorado; Department of Craniofacial Biology, University of Colorado School of Dental Medicine, Aurora, Colorado (L.E.H.); Immune Disease Institute, Harvard Medical School, Boston, Massachusetts (H.L.); and La Jolla Institute for Allergy and Immunology, La Jolla, California (P.G.H.)
| | - Lynn E Heasley
- Department of Pharmacology (E.S.G., K.M.W., M.L.D.) and Department of Medicine, Division of Renal Diseases and Hypertension (R.A.N.), University of Colorado School of Medicine, Aurora, Colorado; Department of Craniofacial Biology, University of Colorado School of Dental Medicine, Aurora, Colorado (L.E.H.); Immune Disease Institute, Harvard Medical School, Boston, Massachusetts (H.L.); and La Jolla Institute for Allergy and Immunology, La Jolla, California (P.G.H.)
| | - Mark L Dell'Acqua
- Department of Pharmacology (E.S.G., K.M.W., M.L.D.) and Department of Medicine, Division of Renal Diseases and Hypertension (R.A.N.), University of Colorado School of Medicine, Aurora, Colorado; Department of Craniofacial Biology, University of Colorado School of Dental Medicine, Aurora, Colorado (L.E.H.); Immune Disease Institute, Harvard Medical School, Boston, Massachusetts (H.L.); and La Jolla Institute for Allergy and Immunology, La Jolla, California (P.G.H.)
| |
Collapse
|
9
|
Wang YC, Cui XB, Chuang YH, Chen SY. Janus Kinase 3, a Novel Regulator for Smooth Muscle Proliferation and Vascular Remodeling. Arterioscler Thromb Vasc Biol 2017; 37:1352-1360. [PMID: 28473442 DOI: 10.1161/atvbaha.116.308895] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 04/25/2017] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Vascular remodeling because of smooth muscle cell (SMC) proliferation is a common process occurring in several vascular diseases, such as atherosclerosis, aortic aneurysm, post-transplant vasculopathy, restenosis after angioplasty, etc. The molecular mechanism underlying SMC proliferation, however, is not completely understood. The objective of this study is to determine the role and mechanism of Janus kinase 3 (JAK3) in vascular remodeling and SMC proliferation. APPROACH AND RESULTS Platelet-derived growth factor-BB, an SMC mitogen, induces JAK3 expression and phosphorylation while stimulating SMC proliferation. Janex-1, a specific inhibitor of JAK3, or knockdown of JAK3 by short hairpin RNA, inhibits the SMC proliferation. Conversely, ectopic expression of JAK3 promotes SMC proliferation. Mechanistically, JAK3 promotes the phosphorylation of signal transducer and activator of transcription 3 and c-Jun N-terminal kinase in SMC, 2 signaling pathways known to be critical for SMC proliferation and vascular remodeling. Blockade of these 2 signaling pathways by their inhibitors impeded the JAK3-mediated SMC proliferation. In vivo, knockdown of JAK3 attenuates injury-induced neointima formation with attenuated neointimal SMC proliferation. Knockdown of JAK3 also induces neointimal SMC apoptosis in rat carotid artery balloon injury model. CONCLUSIONS Our results demonstrate that JAK3 mediates SMC proliferation and survival during injury-induced vascular remodeling, which provides a potential therapeutic target for preventing neointimal hyperplasia in proliferative vascular diseases.
Collapse
Affiliation(s)
- Yung-Chun Wang
- From the Department of Physiology and Pharmacology, University of Georgia, Athens
| | - Xiao-Bing Cui
- From the Department of Physiology and Pharmacology, University of Georgia, Athens
| | - Ya-Hui Chuang
- From the Department of Physiology and Pharmacology, University of Georgia, Athens
| | - Shi-You Chen
- From the Department of Physiology and Pharmacology, University of Georgia, Athens.
| |
Collapse
|
10
|
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
|
11
|
Frismantiene A, Dasen B, Pfaff D, Erne P, Resink TJ, Philippova M. T-cadherin promotes vascular smooth muscle cell dedifferentiation via a GSK3β-inactivation dependent mechanism. Cell Signal 2016; 28:516-530. [DOI: 10.1016/j.cellsig.2016.02.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/12/2016] [Accepted: 02/18/2016] [Indexed: 11/24/2022]
|
12
|
Nuclear PTEN functions as an essential regulator of SRF-dependent transcription to control smooth muscle differentiation. Nat Commun 2016; 7:10830. [PMID: 26940659 PMCID: PMC5411712 DOI: 10.1038/ncomms10830] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 01/25/2016] [Indexed: 12/13/2022] Open
Abstract
Vascular disease progression is associated with marked changes in vascular smooth muscle cell (SMC) phenotype and function. SMC contractile gene expression and, thus differentiation, is under direct transcriptional control by the transcription factor, serum response factor (SRF); however, the mechanisms dynamically regulating SMC phenotype are not fully defined. Here we report that the lipid and protein phosphatase, PTEN, has a novel role in the nucleus by functioning as an indispensible regulator with SRF to maintain the differentiated SM phenotype. PTEN interacts with the N-terminal domain of SRF and PTEN–SRF interaction promotes SRF binding to essential promoter elements in SM-specific genes. Factors inducing phenotypic switching promote loss of nuclear PTEN through nucleo-cytoplasmic translocation resulting in reduced myogenically active SRF, but enhanced SRF activity on target genes involved in proliferation. Overall decreased expression of PTEN was observed in intimal SMCs of human atherosclerotic lesions underlying the potential clinical importance of these findings. The transcription factor, serum response factor, SRF regulates critical smooth muscle (SM) contractile gene expression but what else controls SM differentiation is unclear. Here, Horita et al. demonstrate that nuclear PTEN acts with SRF at the transcriptional level to maintain the differentiated SM phenotype.
Collapse
|
13
|
Lupieri A, Smirnova N, Malet N, Gayral S, Laffargue M. PI3K signaling in arterial diseases: Non redundant functions of the PI3K isoforms. Adv Biol Regul 2015; 59:4-18. [PMID: 26238239 DOI: 10.1016/j.jbior.2015.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 06/15/2015] [Accepted: 06/15/2015] [Indexed: 06/04/2023]
Abstract
Cardiovascular diseases are the most common cause of death around the world. This includes atherosclerosis and the adverse effects of its treatment, such as restenosis and thrombotic complications. The development of these arterial pathologies requires a series of highly-intertwined interactions between immune and arterial cells, leading to specific inflammatory and fibroproliferative cellular responses. In the last few years, the study of phosphoinositide 3-kinase (PI3K) functions has become an attractive area of investigation in the field of arterial diseases, especially since inhibitors of specific PI3K isoforms have been developed. The PI3K family includes 8 members divided into classes I, II or III depending on their substrate specificity. Although some of the different isoforms are responsible for the production of the same 3-phosphoinositides, they each have specific, non-redundant functions as a result of differences in expression levels in different cell types, activation mechanisms and specific subcellular locations. This review will focus on the functions of the different PI3K isoforms that are suspected as having protective or deleterious effects in both the various immune cells and types of cell found in the arterial wall. It will also discuss our current understanding in the context of which PI3K isoform(s) should be targeted for future therapeutic interventions to prevent or treat arterial diseases.
Collapse
Affiliation(s)
- Adrien Lupieri
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Natalia Smirnova
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Nicole Malet
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Stéphanie Gayral
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France
| | - Muriel Laffargue
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, F-31300, France.
| |
Collapse
|
14
|
Guo X, Shi N, Cui XB, Wang JN, Fukui Y, Chen SY. Dedicator of cytokinesis 2, a novel regulator for smooth muscle phenotypic modulation and vascular remodeling. Circ Res 2015; 116:e71-80. [PMID: 25788409 DOI: 10.1161/circresaha.116.305863] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 03/18/2015] [Indexed: 12/20/2022]
Abstract
RATIONALE Vascular smooth muscle cell (SMC) phenotypic modulation and vascular remodeling contribute to the development of several vascular disorders such as restenosis after angioplasty, transplant vasculopathy, and atherosclerosis. The mechanisms underlying these processes, however, remain largely unknown. OBJECTIVE The objective of this study is to determine the role of dedicator of cytokinesis 2 (DOCK2) in SMC phenotypic modulation and vascular remodeling. METHODS AND RESULTS Platelet-derived growth factor-BB induced DOCK2 expression while modulating SMC phenotype. DOCK2 deficiency diminishes platelet-derived growth factor-BB or serum-induced downregulation of SMC markers. Conversely, DOCK2 overexpression inhibits SMC marker expression in primary cultured SMC. Mechanistically, DOCK2 inhibits myocardin expression, blocks serum response factor nuclear location, attenuates myocardin binding to serum response factor, and thus attenuates myocardin-induced smooth muscle marker promoter activity. Moreover, DOCK2 and Kruppel-like factor 4 cooperatively inhibit myocardin-serum response factor interaction. In a rat carotid artery balloon-injury model, DOCK2 is induced in media layer SMC initially and neointima SMC subsequently after vascular injury. Knockdown of DOCK2 dramatically inhibits the neointima formation by 60%. Most importantly, knockout of DOCK2 in mice markedly blocks ligation-induced intimal hyperplasia while restoring SMC contractile protein expression. CONCLUSIONS Our studies identified DOCK2 as a novel regulator for SMC phenotypic modulation and vascular lesion formation after vascular injury. Therefore, targeting DOCK2 may be a potential therapeutic strategy for the prevention of vascular remodeling in proliferative vascular diseases.
Collapse
Affiliation(s)
- Xia Guo
- From the Department of Physiology and Pharmacology, University of Georgia, Athens (X.G., N.S., X.-B.C., S.-Y.C.); Department of Cardiology, Institute of Clinical Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China (J.-N.W., S.-Y.C.); and Department of Immunobiology and Neuroscience, Kyushu University, Fukuoka, Japan (Y.F.)
| | - Ning Shi
- From the Department of Physiology and Pharmacology, University of Georgia, Athens (X.G., N.S., X.-B.C., S.-Y.C.); Department of Cardiology, Institute of Clinical Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China (J.-N.W., S.-Y.C.); and Department of Immunobiology and Neuroscience, Kyushu University, Fukuoka, Japan (Y.F.)
| | - Xiao-Bing Cui
- From the Department of Physiology and Pharmacology, University of Georgia, Athens (X.G., N.S., X.-B.C., S.-Y.C.); Department of Cardiology, Institute of Clinical Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China (J.-N.W., S.-Y.C.); and Department of Immunobiology and Neuroscience, Kyushu University, Fukuoka, Japan (Y.F.)
| | - Jia-Ning Wang
- From the Department of Physiology and Pharmacology, University of Georgia, Athens (X.G., N.S., X.-B.C., S.-Y.C.); Department of Cardiology, Institute of Clinical Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China (J.-N.W., S.-Y.C.); and Department of Immunobiology and Neuroscience, Kyushu University, Fukuoka, Japan (Y.F.)
| | - Yoshinori Fukui
- From the Department of Physiology and Pharmacology, University of Georgia, Athens (X.G., N.S., X.-B.C., S.-Y.C.); Department of Cardiology, Institute of Clinical Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China (J.-N.W., S.-Y.C.); and Department of Immunobiology and Neuroscience, Kyushu University, Fukuoka, Japan (Y.F.)
| | - Shi-You Chen
- From the Department of Physiology and Pharmacology, University of Georgia, Athens (X.G., N.S., X.-B.C., S.-Y.C.); Department of Cardiology, Institute of Clinical Medicine, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China (J.-N.W., S.-Y.C.); and Department of Immunobiology and Neuroscience, Kyushu University, Fukuoka, Japan (Y.F.).
| |
Collapse
|
15
|
The transcription factor serum response factor stimulates axon regeneration through cytoplasmic localization and cofilin interaction. J Neurosci 2014; 33:18836-48. [PMID: 24285890 DOI: 10.1523/jneurosci.3029-13.2013] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Axonal injury generates growth inert retraction bulbs with dynamic cytoskeletal properties that are severely compromised. Conversion of "frozen" retraction bulbs into actively progressing growth cones is a major aim in axon regeneration. Here we report that murine serum response factor (SRF), a gene regulator linked to the actin cytoskeleton, modulates growth cone actin dynamics during axon regeneration. In regeneration-competent facial motoneurons, Srf deletion inhibited axonal regeneration. In wild-type mice after nerve injury, SRF translocated from the nucleus to the cytoplasm, suggesting a cytoplasmic SRF function in axonal regeneration. Indeed, adenoviral overexpression of cytoplasmic SRF (SRF-ΔNLS-GFP) stimulated axonal sprouting and facial nerve regeneration in vivo. In primary central and peripheral neurons, SRF-ΔNLS-GFP stimulated neurite outgrowth, branch formation, and growth cone morphology. Furthermore, we uncovered a link between SRF and the actin-severing factor cofilin during axonal regeneration in vivo. Facial nerve axotomy increased the total cofilin abundance and also nuclear localization of phosphorylated cofilin in a subpopulation of lesioned motoneurons. This cytoplasmic-to-nucleus translocation of P-cofilin upon axotomy was reduced in motoneurons expressing SRF-ΔNLS-GFP. Finally, we demonstrate that cytoplasmic SRF and cofilin formed a reciprocal regulatory unit. Overexpression of cytoplasmic SRF reduced cofilin phosphorylation and vice versa: overexpression of cofilin inhibited SRF phosphorylation. Therefore, a regulatory loop consisting of SRF and cofilin might take part in reactivating actin dynamics in growth-inert retraction bulbs and facilitating axon regeneration.
Collapse
|
16
|
Fan Z, Li C, Qin C, Xie L, Wang X, Gao Z, Qiangbacuozhen, Wang T, Yu L, Liu H. Role of the PI3K/AKT pathway in modulating cytoskeleton rearrangements and phenotype switching in rat pulmonary arterial vascular smooth muscle cells. DNA Cell Biol 2013; 33:12-9. [PMID: 24283363 DOI: 10.1089/dna.2013.2022] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Pulmonary arterial smooth muscle cell (PASMC) phenotype switching, which is characterized by changes in smooth muscle (SM)-specific gene expression, contributes to vascular remodeling in pulmonary hypertension. In addition, it has been shown that the transcription of SM-specific genes is modulated by cytoskeleton rearrangement. However, the intracellular mechanisms and signaling pathways that regulate these relationships are largely unknown. In the present study, we aimed to investigate the roles that phosphoinositide 3-kinase (PI3K) and protein kinase B (PKB), also known as AKT, play in modulating the cytoskeleton and phenotype of rat PASMCs. To observe the downstream effects of inhibiting or enhancing PI3K/AKT pathway activity, we used various approaches to manipulate protein function and gene expression. Treatment of PASMCs with platelet-derived growth factor (PDGF)-BB or PIK3CA-adenovirus induced cytoskeleton rearrangements and downregulated SM22α and α-SM actin gene expression. Inhibition of PI3K led to blocking of AKT phosphorylation and attenuated the PDGF-BB-induced downregulation of F-actin and SM-specific genes, the downstream effector of PI3K. The decrease in SM22α and α-SM actin mRNA levels induced by PDGF-BB was markedly and reproducibly blocked by LY294002. PI3K/AKT pathway plays a vital role in the modulation of PASMCs cytoskeleton rearrangement and phenotype switching.
Collapse
Affiliation(s)
- Zhiyu Fan
- 1 Pulmonary Vascular Remodeling Research Unit, West China Institute of Women's and Children's Health, West China Second University Hospital, Sichuan University , Chengdu, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Liu R, Jin Y, Tang WH, Qin L, Zhang X, Tellides G, Hwa J, Yu J, Martin KA. Ten-eleven translocation-2 (TET2) is a master regulator of smooth muscle cell plasticity. Circulation 2013; 128:2047-57. [PMID: 24077167 DOI: 10.1161/circulationaha.113.002887] [Citation(s) in RCA: 212] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Smooth muscle cells (SMCs) are remarkably plastic. Their reversible differentiation is required for growth and wound healing but also contributes to pathologies such as atherosclerosis and restenosis. Although key regulators of the SMC phenotype, including myocardin (MYOCD) and KLF4, have been identified, a unifying epigenetic mechanism that confers reversible SMC differentiation has not been reported. METHODS AND RESULTS Using human SMCs, human arterial tissue, and mouse models, we report that SMC plasticity is governed by the DNA-modifying enzyme ten-eleven translocation-2 (TET2). TET2 and its product, 5-hydroxymethylcytosine (5-hmC), are enriched in contractile SMCs but reduced in dedifferentiated SMCs. TET2 knockdown inhibits expression of key procontractile genes, including MYOCD and SRF, with concomitant transcriptional upregulation of KLF4. TET2 knockdown prevents rapamycin-induced SMC differentiation, whereas TET2 overexpression is sufficient to induce a contractile phenotype. TET2 overexpression also induces SMC gene expression in fibroblasts. Chromatin immunoprecipitation demonstrates that TET2 coordinately regulates phenotypic modulation through opposing effects on chromatin accessibility at the promoters of procontractile versus dedifferentiation-associated genes. Notably, we find that TET2 binds and 5-hmC is enriched in CArG-rich regions of active SMC contractile promoters (MYOCD, SRF, and MYH11). Loss of TET2 and 5-hmC positively correlates with the degree of injury in murine models of vascular injury and human atherosclerotic disease. Importantly, localized TET2 knockdown exacerbates injury response, and local TET2 overexpression restores the 5-hmC epigenetic landscape and contractile gene expression and greatly attenuates intimal hyperplasia in vivo. CONCLUSIONS We identify TET2 as a novel and necessary master epigenetic regulator of SMC differentiation.
Collapse
Affiliation(s)
- Renjing Liu
- Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (R.L., Y.J., W.T., X.Z., J.H., J.Y., K.A.M.), Department of Surgery (Cardiac Surgery) (L.Q., G.T.), and Department of Pharmacology (K.A.M.), Yale University, New Haven, CT
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Dipaolo BC, Davidovich N, Kazanietz MG, Margulies SS. Rac1 pathway mediates stretch response in pulmonary alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol 2013; 305:L141-53. [PMID: 23686855 DOI: 10.1152/ajplung.00298.2012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Alveolar epithelial cells (AECs) maintain the pulmonary blood-gas barrier integrity with gasketlike intercellular tight junctions (TJ) that are anchored internally to the actin cytoskeleton. We have previously shown that AEC monolayers stretched cyclically and equibiaxially undergo rapid magnitude- and frequency-dependent actin cytoskeletal remodeling to form perijunctional actin rings (PJARs). In this work, we show that even 10 min of stretch induced increases in the phosphorylation of Akt and LIM kinase (LIMK) and decreases in cofilin phosphorylation, suggesting that the Rac1/Akt pathway is involved in these stretch-mediated changes. We confirmed that Rac1 inhibitors wortmannin or EHT-1864 decrease stretch-stimulated Akt and LIMK phosphorylation and that Rac1 agonists PIP3 or PDGF increase phosphorylation of these proteins in unstretched cells. We also confirmed that Rac1 pathway inhibition during stretch modulated stretch-induced changes in occludin content and monolayer permeability, actin remodeling and PJAR formation, and cell death. As further validation, overexpression of Rac GTPase-activating protein β2-chimerin also preserved monolayer barrier properties in stretched monolayers. In summary, our data suggest that constitutive activity of Rac1, which is necessary for stretch-induced activation of the Rac1 downstream proteins, mediates stretch-induced increases in permeability and PJAR formation.
Collapse
Affiliation(s)
- Brian C Dipaolo
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
19
|
Guan H, Chen C, Zhu L, Cui C, Guo Y, Fu M, Wang L, Tang Q. Indole-3-carbinol blocks platelet-derived growth factor-stimulated vascular smooth muscle cell function and reduces neointima formation in vivo. J Nutr Biochem 2013; 24:62-9. [DOI: 10.1016/j.jnutbio.2012.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 02/08/2012] [Accepted: 02/08/2012] [Indexed: 02/01/2023]
|
20
|
Ge Q, Moir LM, Trian T, Niimi K, Poniris M, Shepherd PR, Black JL, Oliver BG, Burgess JK. The phosphoinositide 3'-kinase p110δ modulates contractile protein production and IL-6 release in human airway smooth muscle. J Cell Physiol 2012; 227:3044-52. [PMID: 22015454 DOI: 10.1002/jcp.23046] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Transforming growth factor (TGF) β1 increases pro-inflammatory cytokines and contractile protein expression by human airway smooth muscle (ASM) cells, which could augment airway inflammation and hyperresponsiveness. Phosphoinositide 3' kinase (PI3K) is one of the signaling pathways implicated in TGFβ1 stimulation, and may be altered in asthmatic airways. This study compared the expression of PI3K isoforms by ASM cells from donors with asthma (A), chronic obstructive pulmonary disease (COPD), or neither disease (NA), and investigated the role of PI3K isoforms in the production of TGFβ1 induced pro-inflammatory cytokine and contractile proteins in ASM cells. A cells expressed higher basal levels of p110δ mRNA compared to NA and COPD cells; however COPD cells produced more p110δ protein. TGFβ1 increased 110δ mRNA expression to the same extent in the three groups. Neither the p110δ inhibitor IC87114 (1, 10, 30 µM), the p110β inhibitor TGX221 (0.1, 1, 10 µM) nor the PI3K pan inhibitor LY294002 (3, 10 µM) had any effect on basal IL-6, calponin or smooth muscle α-actin (α-SMA) expression. However, TGFβ1 increased calponin and α-SMA expression was inhibited by IC87114 and LY294002 in all three groups. IC87114, TGX221, and LY294002 reduced TGFβ1 induced IL-6 release in a dose related manner in all groups of ASM cells. PI3K p110δ is important for TGFβ1 induced production of the contractile proteins calponin and α-SMA and the proinflammatory cytokine IL-6 in ASM cells, and may therefore be relevant as a potential therapeutic target to treat both inflammation and airway remodeling.
Collapse
Affiliation(s)
- Qi Ge
- Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Horita HN, Simpson PA, Ostriker A, Furgeson S, Van Putten V, Weiser-Evans MCM, Nemenoff RA. Serum response factor regulates expression of phosphatase and tensin homolog through a microRNA network in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 2011; 31:2909-19. [PMID: 21940949 DOI: 10.1161/atvbaha.111.233585] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Serum response factor (SRF) is a critical transcription factor in smooth muscle cells (SMCs) controlling differentiation and proliferation. Our previous work demonstrated that depleting SRF in cultured SMCs decreased expression of SMC markers but increased proliferation and inflammatory mediators. A similar phenotype has been observed in SMCs silenced for phosphatase and tensin homolog (PTEN), suggesting that SRF and PTEN may lie on a common pathway. Our goal was to determine the effect of SRF depletion on PTEN levels and define mechanisms mediating this effect. METHODS AND RESULTS In SRF-silenced SMCs, PTEN protein levels but not mRNA levels were decreased, suggesting posttranscriptional regulation. Reintroduction of PTEN into SRF-depleted SMCs reversed increases in proliferation and cytokine/chemokine production but had no effect on SMC marker expression. SRF-depleted cells showed decreased levels of microRNA (miR)-143 and increased miR-21, which was sufficient to suppress PTEN. Increased miR-21 expression was dependent on induction of Fos related antigen (FRA)-1, which is a direct target of miR-143. Introducing miR-143 into SRF-depleted SMCs reduced FRA-1 expression and miR-21 levels and restored PTEN expression. CONCLUSIONS SRF regulates PTEN expression in SMCs through a miR network involving miR-143, targeting FRA-1, which regulates miR-21. Cross-talk between SRF and PTEN likely represents a critical axis in phenotypic remodeling of SMCs.
Collapse
Affiliation(s)
- Henrick N Horita
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Desai LP, Wu Y, Tepper RS, Gunst SJ. Mechanical stimuli and IL-13 interact at integrin adhesion complexes to regulate expression of smooth muscle myosin heavy chain in airway smooth muscle tissue. Am J Physiol Lung Cell Mol Physiol 2011; 301:L275-84. [PMID: 21642449 DOI: 10.1152/ajplung.00043.2011] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Airway smooth muscle phenotype may be modulated in response to external stimuli under physiological and pathophysiological conditions. The effect of mechanical forces on airway smooth muscle phenotype were evaluated in vitro by suspending weights of 0.5 or 1 g from the ends of canine tracheal smooth muscle tissues, incubating the weighted tissues for 6 h, and then measuring the expression of the phenotypic marker protein, smooth muscle myosin heavy chain (SmMHC). Incubation of the tissues at a high load significantly increased expression of SmMHC compared with incubation at low load. Incubation of the tissues at a high load also decreased activation of PKB/Akt, as indicated by its phosphorylation at Ser 473. Inhibition of Akt or phosphatidylinositol-3,4,5 triphosphate-kinase increased SmMHC expression in tissues at low load but did not affect SmMHC expression at high load. IL-13 induced a significant increase in Akt activation and suppressed the expression of SmMHC protein at both low and high loads. The role of integrin signaling in mechanotransduction was evaluated by expressing a PINCH (LIM1-2) fragment in the muscle tissues that prevents the membrane localization of the integrin-binding IPP complex (ILK/PINCH/α-parvin), and also by expressing an inactive integrin-linked kinase mutant (ILK S343A) that inhibits endogenous ILK activity. Both mutants inhibited Akt activation and increased expression of SmMHC protein at low load but had no effect at high load. These results suggest that mechanical stress and IL-13 both act through an integrin-mediated signaling pathway to oppositely regulate the expression of phenotypic marker proteins in intact airway smooth muscle tissues. The stimulatory effects of mechanical stress on contractile protein expression oppose the suppression of contractile protein expression mediated by IL-13; thus the imposition of mechanical strain may inhibit changes in airway smooth muscle phenotype induced by inflammatory mediators.
Collapse
Affiliation(s)
- Leena P Desai
- Dept. of Cellular & Integrative Physiology, Indiana Univ. School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | |
Collapse
|
23
|
Ma L, Brown M, Kogut P, Serban K, Li X, McConville J, Chen B, Bentley JK, Hershenson MB, Dulin N, Solway J, Camoretti-Mercado B. Akt activation induces hypertrophy without contractile phenotypic maturation in airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2011; 300:L701-9. [PMID: 21378028 DOI: 10.1152/ajplung.00119.2009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Airway smooth muscle (ASM) hypertrophy is a cardinal feature of severe asthma, but the underlying molecular mechanisms remain uncertain. Forced protein kinase B/Akt 1 activation is known to induce myocyte hypertrophy in other muscle types, and, since a number of mediators present in asthmatic airways can activate Akt signaling, we hypothesized that Akt activation could contribute to ASM hypertrophy in asthma. To test this hypothesis, we evaluated whether Akt activation occurs naturally within airway myocytes in situ, whether Akt1 activation is sufficient to cause hypertrophy of normal airway myocytes, and whether such hypertrophy is accompanied by excessive accumulation of contractile apparatus proteins (contractile phenotype maturation). Immunostains of human airway sections revealed concordant activation of Akt (reflected in Ser(473) phosphorylation) and of its downstream effector p70(S6Kinase) (reflected in Thr(389) phosphorylation) within airway muscle bundles, but there was no phosphorylation of the alternative Akt downstream target glycogen synthase kinase (GSK) 3β. Artificial overexpression of constitutively active Akt1 (by plasmid transduction or lentiviral infection) caused a progressive increase in size and protein content of cultured canine tracheal myocytes and increased p70(S6Kinase) phosphorylation but not GSK3β phosphorylation; however, constitutively active Akt1 did not cause disproportionate overaccumulation of smooth muscle (sm) α-actin and SM22. Furthermore, mRNAs encoding sm-α-actin and SM22 were reduced. These results indicate that forced Akt1 signaling causes hypertrophy of cultured airway myocytes without inducing further contractile phenotypic maturation, possibly because of opposing effects on contractile protein gene transcription and translation, and suggest that natural activation of Akt1 plays a similar role in asthmatic ASM.
Collapse
Affiliation(s)
- Lan Ma
- Department of Medicine, University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Zhao Y, Biswas SK, McNulty PH, Kozak M, Jun JY, Segar L. PDGF-induced vascular smooth muscle cell proliferation is associated with dysregulation of insulin receptor substrates. Am J Physiol Cell Physiol 2011; 300:C1375-85. [PMID: 21325637 DOI: 10.1152/ajpcell.00670.2008] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In vascular smooth muscle cells (VSMCs), platelet-derived growth factor (PDGF) plays a major role in inducing phenotypic switching from contractile to proliferative state. Importantly, VSMC phenotypic switching is also determined by the phosphorylation state/expression levels of insulin receptor substrate (IRS), an intermediary signaling component that is shared by insulin and IGF-I. To date, the roles of PDGF-induced key proliferative signaling components including Akt, p70S6kinase, and ERK1/2 on the serine phosphorylation/expression of IRS-1 and IRS-2 isoforms remain unclear in VSMCs. We hypothesize that PDGF-induced VSMC proliferation is associated with dysregulation of insulin receptor substrates. Using human aortic VSMCs, we demonstrate that prolonged PDGF treatment led to sustained increases in the phosphorylation of protein kinases such as Akt, p70S6kinase, and ERK1/2, which mediate VSMC proliferation. In addition, PDGF enhanced IRS-1/IRS-2 serine phosphorylation and downregulated IRS-2 expression in a time- and concentration-dependent manner. Notably, phosphoinositide 3-kinase (PI 3-kinase) inhibitor (PI-103) and mammalian target of rapamycin inhibitor (rapamycin), which abolished PDGF-induced Akt and p70S6kinase phosphorylation, respectively, blocked PDGF-induced IRS-1 serine phosphorylation and IRS-2 downregulation. In contrast, MEK1/ERK inhibitor (U0126) failed to block PDGF-induced IRS-1 serine phosphorylation and IRS-2 downregulation. PDGF-induced IRS-2 downregulation was prevented by lactacystin, an inhibitor of proteasomal degradation. Functionally, PDGF-mediated IRS-1/IRS-2 dysregulation resulted in the attenuation of insulin-induced IRS-1/IRS-2-associated PI 3-kinase activity. Pharmacological inhibition of PDGF receptor tyrosine kinase with imatinib prevented IRS-1/IRS-2 dysregulation and restored insulin receptor signaling. In conclusion, strategies to inhibit PDGF receptors would not only inhibit neointimal growth but may provide new therapeutic options to prevent dysregulated insulin receptor signaling in VSMCs in nondiabetic and diabetic states.
Collapse
Affiliation(s)
- Yan Zhao
- Departments of Medicine and Pharmacology, Heart & Vascular Institute, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | | | | | | | | | | |
Collapse
|
25
|
McConville JF, Fernandes DJ, Churchill J, Dewundara S, Kogut P, Shah S, Fuchs G, Kedainis D, Bellam SK, Patel NM, McCauley J, Dulin NO, Gupta MP, Adam S, Yoneda Y, Camoretti-Mercado B, Solway J. Nuclear import of serum response factor in airway smooth muscle. Am J Respir Cell Mol Biol 2010; 45:453-8. [PMID: 21131446 DOI: 10.1165/rcmb.2008-0393oc] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We have previously shown that the transcription-promoting activity of serum response factor (SRF) is partially regulated by its extranuclear redistribution. In this study, we examined the cellular mechanisms that facilitate SRF nuclear entry in canine tracheal smooth muscle cells. We used in vitro pull-down assays to determine which karyopherin proteins bound SRF and found that SRF binds KPNA1 and KPNB1 through its nuclear localization sequence. Immunoprecipitation studies also demonstrated direct SRF-KPNA1 interaction in HEK293 cells. Import assays demonstrated that KPNA1 and KPNB1 together were sufficient to mediate rapid nuclear import of SRF-GFP. Our studies also suggest that SRF is able to gain nuclear entry through an auxiliary, nuclear localization sequence-independent mechanism.
Collapse
Affiliation(s)
- John F McConville
- Department of Medicine, University of Chicago, 5841 S. Maryland Avenue, MC6026, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Chronic treatment with PDGF-BB and endothelin-1 synergistically induces vascular hyperplasia and loss of contractility in organ-cultured rat tail artery. Atherosclerosis 2010; 214:288-94. [PMID: 21129745 DOI: 10.1016/j.atherosclerosis.2010.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2009] [Revised: 10/23/2010] [Accepted: 11/04/2010] [Indexed: 01/21/2023]
Abstract
OBJECTIVE In this study, we examined the synergistic effects of the two potent pathogenic factors, platelet-derived growth factor-BB (PDGF-BB) and endothelin-1 (ET-1) to induce vascular hyperplasia using ex vivo organ-culture system. METHODS AND RESULTS In organ-cultured rat tail arteries, concomitant treatment with 100 ng/ml PDGF-BB and 300 nM ET-1 for 4 days induced medial hyperplasia with increased smooth muscle cell proliferation. Concomitant treatment with PDGF-BB (10-300 nM) and ET-1 (30 nM-1 μM) dose-dependently suppressed contractile responses to high K(+) and norepinephrine. This dyscontractility was accompanied by decreased α-actin protein expression. In all series of experiments, concomitant treatment with PDGF-BB and ET-1 exhibited stronger effects than sole treatment with PDGF-BB (100 ng/ml) or ET-1 (300 nM). Western blot analysis revealed that concomitant treatment with PDGF-BB and ET-1 synergistically phosphorylated extracellular signal-regulated kinase 1 and 2 (ERK1/2), Akt, and a downstream target of mammalian target of rapamycin (mTOR), p70 ribosomal S6 kinase in cultured artery. Consistently, a MAPK/ERK kinase (MEK) inhibitor, PD98059 (30 μM), a phosphoinositide 3-kinase (PI3K) inhibitor, LY294002, and an mTOR inhibitor, rapamycin (30 nM), partially restored PDGF-BB and ET-1-induced hyperplastic changes. CONCLUSIONS We evidenced for the first time at tissue level that PDGF-BB and ET-1 synergistically accelerate vascular smooth muscle hyperplastic changes and lose its contractility, at least partially through ERK1/2, Akt, and mTOR activation.
Collapse
|
27
|
Jie W, Guo J, Shen Z, Wang X, Zheng S, Wang G, Ao Q. Contribution of myocardin in the hypoxia-induced phenotypic switching of rat pulmonary arterial smooth muscle cells. Exp Mol Pathol 2010; 89:301-6. [PMID: 20621093 DOI: 10.1016/j.yexmp.2010.06.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 06/27/2010] [Accepted: 06/29/2010] [Indexed: 12/19/2022]
Abstract
BACKGROUND Hypoxic exposure contributes to the phenotypic switching of smooth muscle cells (SMCs), while the mechanisms involved in this process is not yet fully elucidated. Myocardin as a co-actor of serum reaction factor plays a crucial role in differentiation of SMCs. This study was aimed to investigate the role of myocardin in hypoxia-induced phenotypic switching of rat pulmonary arterial SMCs (PASMCs). METHODS Primary PASMCs were cultured under normoxia and hypoxia (3%O(2), 48 h) respectively, and then the cell proliferation was assessed and the expression of SM22α, osteopontin (contractile and synthetic marker of SMCs, respectively), myocardin and platelet-derived growth factor-BB (PDGF-BB) were detected. After pGCSIL-GFP-shMYOCD lentviral vector was transduced to the PASMCs, the expression of myocardin and SM22α were examined. Moreover, myocardin expression in PASMCs treated with medium enriched with PDGF-BB and conditional medium (CM) from normoxia- and hypoxia-exposed PASMCs was assessed. RESULTS Exposing PASMCs to hypoxia led to an increased cell numbers and the up-regulation of proliferating cell nuclear antigen (PCNA), osteopontin and PDGF-BB; moreover, a significant down-regulation of SM22α and myocardin was identified. Further analysis revealed that knock-down of myocardin with pGCSIL-GFP-shMYOCD vector followed by a decreased SM22α in the PASMCs, and treatment of PASMCs with either exogenous PDGF-BB or hypoxic CM led to a marked decrease of myocardin. CONCLUSIONS Our findings suggest that the decrease of myocardin in PASMCs exposed to hypoxia is partly regulated by the increase of PDGF-BB, which contributes to the phenotypic switching of PASMCs in hypoxic condition.
Collapse
Affiliation(s)
- Wei Jie
- Institute of Pathology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Smooth muscle cells (SMCs) retain remarkable plasticity to undergo phenotypic modulation in which the expression of smooth muscle markers is markedly attenuated while conversely expression of extracellular matrix (ECM) is dramatically up-regulated. Myocardin is perhaps the most potent transcription factor for stimulating expression of smooth muscle-specific genes; little is known, however, about whether myocardin can orchestrate ECM expression to act in concert with smooth muscle differentiation program. In this study, we demonstrated myocardin coordinate smooth muscle differentiation by inducing transcription of microRNA-143 (miR-143), which attenuates ECM versican protein expression. Previous studies have shown that versican is a chondroitin sulfate proteoglycan of the ECM that is produced by synthetic SMCs and promotes SMC migration and proliferation. Our data demonstrated that myocardin significantly represses versican expression in multiple cell lines, and this occurs through the induction of miR-143 by myocardin. By a modified reverse transcribed PCR, we found that miR-143 specifically binds to the 3'-untranslated region of versican mRNA. Reporter assays validated that miR-143 targets versican 3'-untranslated region through an evolutionarily conserved miR-143 binding site. Furthermore, overexpression of miR-143 significantly represses versican expression, whereas conversely, depletion of endogenous miR-143 results in up-regulation of versican expression. In addition, we demonstrated that myocardin represses versican through induction of miR-143. Finally, we found that the regulation of versican by miR-143 is involved in platelet-derived growth factor BB-induced SMC migration. This study provides the first evidence that myocardin, in addition to activating smooth muscle-specific genes, regulates ECM expression through induction of microRNAs during smooth muscle differentiation.
Collapse
Affiliation(s)
- Xiaobo Wang
- Center for Cardiovascular Sciences, Albany Medical College, Albany, New York 12208, USA
| | | | | |
Collapse
|
29
|
Furgeson SB, Simpson PA, Park I, Vanputten V, Horita H, Kontos CD, Nemenoff RA, Weiser-Evans MCM. Inactivation of the tumour suppressor, PTEN, in smooth muscle promotes a pro-inflammatory phenotype and enhances neointima formation. Cardiovasc Res 2010; 86:274-82. [PMID: 20051384 DOI: 10.1093/cvr/cvp425] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS Phosphatase and tensin homolog (PTEN) is implicated as a negative regulator of vascular smooth muscle cell (SMC) proliferation and injury-induced vascular remodelling. We tested if selective depletion of PTEN only in SMC is sufficient to promote SMC phenotypic modulation, cytokine production, and enhanced neointima formation. METHODS AND RESULTS Smooth muscle marker expression and induction of pro-inflammatory cytokines were compared in cultured SMC expressing control or PTEN-specific shRNA. Compared with controls, PTEN-deficient SMC exhibited increased phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signalling and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kappaB) activity, reduced expression of SM markers (SM-alpha-actin and calponin), and increased production of stromal cell-derived factor-1alpha (SDF-1alpha), monocyte chemotactic protein-1 (MCP-1), interleukin-6 (IL-6), and chemokine (C-X-C motif) ligand 1 (KC/CXCL1) under basal conditions. PI3K/Akt or mTOR inhibition reversed repression of SM marker expression, whereas PI3K/Akt or NF-kappaB inhibition blocked cytokine induction mediated by PTEN depletion. Carotid ligation in mice with genetic reduction of PTEN specifically in SMC (SMC-specific PTEN heterozygotes) resulted in enhanced neointima formation, increased SMC hyperplasia, reduced SM-alpha-actin and calponin expression, and increased NF-kappaB and cytokine expression compared with wild-types. Lesion formation in SMC-specific heterozygotes was similar to lesion formation in global PTEN heterozygotes, indicating that inactivation of PTEN exclusively in SMC is sufficient to induce considerable increases in neointima formation. CONCLUSION PTEN activation specifically in SMC is a common upstream regulator of multiple downstream events involved in pathological vascular remodelling, including proliferation, de-differentiation, and production of multiple cytokines.
Collapse
Affiliation(s)
- Seth B Furgeson
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Liu Y, Li W, Ye C, Lin Y, Cheang TY, Wang M, Zhang H, Wang S, Zhang L, Wang S. Gambogic Acid Induces G0/G1 Cell Cycle Arrest and Cell Migration Inhibition Via Suppressing PDGF Receptor β Tyrosine Phosphorylation and Rac1 Activity in Rat Aortic Smooth Muscle Cells. J Atheroscler Thromb 2010; 17:901-13. [DOI: 10.5551/jat.3491] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Yong Liu
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University
| | - Wen Li
- Laboratory of Department of Surgery, The First Affiliated Hospital, Sun Yat-sen University
| | - CaiSheng Ye
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University
| | - Ying Lin
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University
| | - Tuck-Yun Cheang
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University
| | - Mian Wang
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University
| | - Hui Zhang
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University
| | - SanMing Wang
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University
| | - LongJuan Zhang
- Laboratory of Department of Surgery, The First Affiliated Hospital, Sun Yat-sen University
| | - ShenMing Wang
- Department of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University
| |
Collapse
|
31
|
Yang XP, Pei ZH, Ren J. MAKING UP OR BREAKING UP: THE TORTUOUS ROLE OF PLATELET-DERIVED GROWTH FACTOR IN VASCULAR AGEING. Clin Exp Pharmacol Physiol 2009; 36:739-47. [DOI: 10.1111/j.1440-1681.2009.05182.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
32
|
Cheng Y, Liu X, Yang J, Lin Y, Xu DZ, Lu Q, Deitch EA, Huo Y, Delphin ES, Zhang C. MicroRNA-145, a novel smooth muscle cell phenotypic marker and modulator, controls vascular neointimal lesion formation. Circ Res 2009; 105:158-66. [PMID: 19542014 DOI: 10.1161/circresaha.109.197517] [Citation(s) in RCA: 543] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Phenotypic modulation of vascular smooth muscle cells (VSMCs) plays a critical role in the pathogenesis of a variety of proliferative vascular diseases. Recently, we have found that microRNA (miRNA) miR-145 is the most abundant miRNA in normal vascular walls and in freshly isolated VSMCs; however, the role of miR-145 in VSMC phenotypic modulation and vascular diseases is currently unknown. Here we find that miR-145 is selectively expressed in VSMCs of the vascular wall and its expression is significantly downregulated in the vascular walls with neointimal lesion formation and in cultured dedifferentiated VSMCs. More importantly, both in cultured rat VSMCs in vitro and in balloon-injured rat carotid arteries in vivo, we demonstrate that the noncoding RNA miR-145 is a novel phenotypic marker and a novel phenotypic modulator of VSMCs. VSMC differentiation marker genes such as SM alpha-actin, calponin, and SM-MHC are upregulated by premiR-145 or adenovirus expressing miR-145 (Ad-miR-145) but are downregulated by the miR-145 inhibitor 2'OMe-miR-145. We have further identified that miR-145-mediated phenotypic modulation of VSMCs is through its target gene KLF5 and its downstream signaling molecule, myocardin. Finally, restoration of miR-145 in balloon-injured arteries via Ad-miR-145 inhibits neointimal growth. We conclude that miR-145 is a novel VSMC phenotypic marker and modulator that is able of controlling vascular neointimal lesion formation. These novel findings may have extensive implications for the diagnosis and therapy of a variety of proliferative vascular diseases.
Collapse
Affiliation(s)
- Yunhui Cheng
- Department of Anesthesiology, New Jersey Medical School, UMDNJ, 185 South Orange Ave, MSB-E548, Newark, NJ 07101, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Functional relevance of tetraspanin CD9 in vascular smooth muscle cell injury phenotypes: A novel target for the prevention of neointimal hyperplasia. Atherosclerosis 2009; 203:377-86. [DOI: 10.1016/j.atherosclerosis.2008.07.036] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Revised: 06/18/2008] [Accepted: 07/22/2008] [Indexed: 11/18/2022]
|
34
|
Wu Y, Huang Y, Herring BP, Gunst SJ. Integrin-linked kinase regulates smooth muscle differentiation marker gene expression in airway tissue. Am J Physiol Lung Cell Mol Physiol 2008; 295:L988-97. [PMID: 18805960 DOI: 10.1152/ajplung.90202.2008] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Phenotypic changes in airway smooth muscle occur with airway inflammation and asthma. These changes may be induced by alterations in the extracellular matrix that initiate signaling pathways mediated by integrin receptors. We hypothesized that integrin-linked kinase (ILK), a multidomain protein kinase that binds to the cytoplasmic tail of beta-integrins, may be an important mediator of signaling pathways that regulate the growth and differentiation state of airway smooth muscle. We disrupted signaling pathways mediated by ILK in intact differentiated tracheal muscle tissues by depleting ILK protein using ILK antisense. The depletion of ILK protein increased the expression of the smooth muscle differentiation marker genes myosin heavy chain (SmMHC), SM22alpha, and calponin and increased the expression of SmMHC protein. Conversely, the overexpression of ILK protein reduced the mRNA levels of SmMHC, SM22alpha, and calponin and SmMHC protein. Analysis by chromatin immunoprecipitation showed that the binding of the transcriptional regulator serum response factor (SRF) to the promoters of SmMHC, SM22alpha, and calponin genes was increased in ILK-depleted tissues and decreased in tissues overexpressing ILK. ILK depletion also increased the amount of SRF that localized within the nucleus. ILK depletion and overexpression, respectively, decreased and increased the activation of its downstream substrate protein kinase B (PKB/Akt). The pharmacological inhibition of Akt activity also increased SRF binding to the promoters of smooth muscle-specific genes and increased expression of smooth muscle proteins, suggesting that ILK may exert its effects by regulating the activity of Akt. We conclude that ILK is a critical regulator of airway smooth muscle differentiation. ILK may mediate signals from integrin receptors that control airway smooth muscle differentiation in response to alterations in the extracellular matrix.
Collapse
Affiliation(s)
- Yidi Wu
- Dept. of Cellular & Integrative Physiology, Indiana Univ. School of Medicine, 635 Barnhill Dr., Indianapolis, IN 46202-5120, USA
| | | | | | | |
Collapse
|
35
|
Hershenson MB, Brown M, Camoretti-Mercado B, Solway J. Airway smooth muscle in asthma. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2008; 3:523-55. [PMID: 18039134 DOI: 10.1146/annurev.pathmechdis.1.110304.100213] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Airway smooth muscle plays a multifaceted role in the pathogenesis of asthma. We review the current understanding of the contribution of airway myocytes to airway inflammation, airway wall remodeling, and airflow obstruction in this prevalent disease syndrome. Together, these roles make airway smooth muscle an attractive target for asthma therapy.
Collapse
Affiliation(s)
- Marc B Hershenson
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | | | |
Collapse
|
36
|
Herrmann J, Haas U, Gressner AM, Weiskirchen R. TGF-β up-regulates serum response factor in activated hepatic stellate cells. Biochim Biophys Acta Mol Basis Dis 2007; 1772:1250-7. [DOI: 10.1016/j.bbadis.2007.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2007] [Revised: 10/17/2007] [Accepted: 10/21/2007] [Indexed: 10/22/2022]
|
37
|
Li N, Zhang J, Liang Y, Shao J, Peng F, Sun M, Xu N, Li X, Wang R, Liu S, Lu Y. A controversial tumor marker: is SM22 a proper biomarker for gastric cancer cells? J Proteome Res 2007; 6:3304-12. [PMID: 17629319 DOI: 10.1021/pr0702363] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
SM22, a dominant protein in smooth muscle cells (SMCs), has been widely reported to be abnormally expressed in many solid tumors. However, the expression patterns of SM22 are not consistent in all tumors, not even in the same ones. Whether SM22 should be considered a tumor biomarker is still debated in different laboratories. Herein, we have carried out a systematical investigation to validate SM22 expression in the primary tissues of gastric cancer (GC). Of eight cases, seven samples were found in the elevated expression of SM22 proteins through proteomic analysis. The observation was further verified by the approaches of Western blotting and quantitative RT-PCR. Surprisingly, the results achieved from tissue microarray in 126 GC cases appeared contrary to the proteomic conclusion, in which the highly expressed SM22 was mainly found in smooth muscle layers, blood vessels, and myofibroblasts. This suggested that the increased abundance of SM22 in the cancerous regions was not caused by the presence of the GC cells. Furthermore, the expression of SM22 was measured in different GC cell lines and SMCs with Western blotting and quantitative RT-PCR. The results revealed that SM22 expression in SMCs was dramatically higher than that of the GC cells, which indicates that SM22 is unlikely to be a proper biomarker for GC. Instead, it can be considered a potential indicator for the abnormal developments of smooth muscles, blood vessels, or myofibroblasts triggered by tumorigenesis.
Collapse
Affiliation(s)
- Na Li
- Beijing Genomics Institute, Chinese Academy of Sciences, Beijing Airport Industrial Zone B-6, Shunyi, Beijing 101318, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Smooth muscle cells (SMCs) possess remarkable phenotypic plasticity that allows rapid adaptation to fluctuating environmental cues. For example, vascular SMCs undergo profound changes in their phenotype during neointimal formation in response to vessel injury or within atherosclerotic plaques. Recent studies have shown that interaction of serum response factor (SRF) and its numerous accessory cofactors with CArG box DNA sequences within promoter chromatin of SMC genes is a nexus for integrating signals that influence SMC differentiation in development and disease. During development, SMC-restricted sets of posttranslational histone modifications are acquired within the CArG box chromatin of SMC genes. These modifications in turn control the chromatin-binding properties of SRF. The histone modifications appear to encode a SMC-specific epigenetic program that is used by extracellular cues to influence SMC differentiation, by regulating binding of SRF and its partners to the chromatin template. Thus, SMC differentiation is dynamically regulated by the interplay between SRF accessory cofactors, the SRF-CArG interaction, and the underlying histone modification program. As such, the inherent plasticity of the SMC lineage offers unique glimpses into how cellular differentiation is dynamically controlled at the level of chromatin within the context of changing microenvironments. Further elucidation of how chromatin regulates SMC differentiation will undoubtedly yield valuable insights into both normal developmental processes and the pathogenesis of several vascular diseases that display detrimental SMC phenotypic behavior.
Collapse
Affiliation(s)
- Oliver G McDonald
- Department of Molecular Physiology and Biological Physics, University of Virginia Health Sciences Center, Charlottesville, VA 22903, USA
| | | |
Collapse
|
39
|
Garin G, Zibara K, Aguilar F, Lo M, Hurlstone A, Poston R, Mcgregor JL. 6A3-5/Osa2 is an early activated gene implicated in the control of vascular smooth muscle cell functions. J Biomed Biotechnol 2007; 2006:97287. [PMID: 17489020 PMCID: PMC1698265 DOI: 10.1155/jbb/2006/97287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Vascular smooth muscle cells (VSMC) growth plays a key role in the pathophysiology of vascular diseases. However, the molecular mechanisms controlling gene transcription in VSMC remain poorly understood. We previously identified, by differential display, a new gene (6A3-5) overexpressed in proliferating rat VSMC. In this study, we have cloned the full-length cDNA by screening a rat foetal brain cDNA library and investigated its functions. The 6A3-5 protein shows 4 putative conserved functional motifs: a DNA binding domain called ARID (AT-rich interaction domain), two recently described motifs (Osa Homology Domain), and a nuclear localization signal. The deduced protein sequence was observed to be 85% identical to the recently described human Osa2 gene. Immunolabelling, using an anti-6A3-5/Osa2 monoclonal antibody, showed a nuclear localization of the 6A3-5/Osa2 protein. In addition, PDGF upregulated 6A3-5/Osa2 expression at both the transcript and protein levels in a dose and time-dependent fashion. The pattern of upregulation by PDGF was reminiscent of the early responsive gene c-fos. The PDGF-induced upregulation of 6A3-5/Osa2 and proliferation of VSMC were significantly inhibited in a dose and sequence-dependent fashion by an antisense, but not by sense, scrambled or mismatched oligonucleotides directed against 6A3-5/Osa2. In VSMC of aortas derived from hypertensive (LH) rats, 6A3-5/Osa2 is overexpressed as compared to that in normotensive (LL) rats. The 6A3-5/Osa2-gene expression is downregulated by an ACE inhibitor and upregulated by exogenous AngiotensinII in LH rats. In summary, these results indicate that 6A3-5/Osa2 is an early activated gene that belongs to a new family of proteins involved in the control of VSMC growth.
Collapse
Affiliation(s)
- Gwenaele Garin
- INSERM XR331, Faculté of Médicine Laënnec, Lyon 69372, France
- Genomics and Atherothrombosis, Thrombosis Research Institute, London SW3 6LR, UK
| | - Kazem Zibara
- INSERM XR331, Faculté of Médicine Laënnec, Lyon 69372, France
- Genomics and Atherothrombosis, Thrombosis Research Institute, London SW3 6LR, UK
| | - Frederick Aguilar
- Département de Physiologie et Pharmacologie Clinique, Faculté de Pharmacie, Université Lyon 1, Lyon, France
| | - Ming Lo
- Département de Physiologie et Pharmacologie Clinique, Faculté de Pharmacie, Université Lyon 1, Lyon, France
| | - Adam Hurlstone
- Hubrecht Laboratory, Netherlands Institute for Developmental Biology, Utrecht, The Netherlands
| | - Robin Poston
- Center for Cardiovascular Biology and Medicine, King's College, University of London, UK
| | - John L. Mcgregor
- INSERM XR331, Faculté of Médicine Laënnec, Lyon 69372, France
- Genomics and Atherothrombosis, Thrombosis Research Institute, London SW3 6LR, UK
- Center for Cardiovascular Biology and Medicine, King's College, University of London, UK
- *John L. Mcgregor:
| |
Collapse
|
40
|
Wang CCL, Sorribas V, Sharma G, Levi M, Draznin B. Insulin attenuates vascular smooth muscle calcification but increases vascular smooth muscle cell phosphate transport. Atherosclerosis 2007; 195:e65-75. [PMID: 17412344 PMCID: PMC2745841 DOI: 10.1016/j.atherosclerosis.2007.02.032] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2006] [Revised: 02/15/2007] [Accepted: 02/23/2007] [Indexed: 12/11/2022]
Abstract
Medial artery vascular smooth muscle cell (VSMC) calcification increases the risk of cardiovascular mortality in type 2 diabetes. However, the influence of insulin on VSMC calcification is unclear. We explored the effects of insulin on rat VSMC calcification in vitro and found that in a dose-dependent fashion, insulin attenuates VSMC calcification induced by high phosphate conditions as quantified by the o-cresolphthalein calcium (OCPC) method. In an in vitro model of insulin resistance in which cells are exposed to elevated insulin concentrations and the PI 3-kinase pathway is selectively inhibited, increased VSMC calcification was observed, suggesting that the PI 3-kinase pathway is involved in this attenuating effect of insulin. We postulated that insulin may also have an effect on phosphate or calcium transport in VSMC. We found that insulin increases phosphate transport at 3 and 24 h. This effect was mediated by increased Vmax for phosphate transport but not Km. Because type III sodium-phosphate co-transporters Pit-1 and Pit-2 are found in VSMC, we examined their expression by Western blot and real-time RT-PCR. Insulin stimulates Pit-1 mRNA modestly (*p<0.01 versus control), an effect inhibited by PD98059 but not by wortmannin. Pit-1 protein expression is induced by insulin, an effect also inhibited by PD98059 (*p<0.001 versus insulin alone). Our results suggest a role for insulin in attenuating VSMC calcification which may be disrupted in selective insulin signaling impairment seen in insulin resistance. This effect of insulin contrasts with its effect to induce phosphate transport in VSMC.
Collapse
Affiliation(s)
- Cecilia C Low Wang
- Research Service, Denver Veterans Affairs Medical Center, Denver, CO 80220, United States.
| | | | | | | | | |
Collapse
|
41
|
de Godoy MAF, Patel CA, Waldman SA, Katsuki M, Regan RF, Rattan S. H-ras inhibits RhoA/ROCK leading to a decrease in the basal tone in the internal anal sphincter. Gastroenterology 2007; 132:1401-9. [PMID: 17408635 DOI: 10.1053/j.gastro.2007.01.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2006] [Accepted: 01/04/2007] [Indexed: 01/13/2023]
Abstract
BACKGROUND & AIMS The present studies evaluated the role of H-ras and its implications in the RhoA/Rho kinase (ROCK) pathway in regulating basal tone in the internal anal sphincter (IAS). METHODS Studies were performed in the IAS from the wild-type (H-ras(+/+)) and knock-out (H-ras(-/-)) mice. The basal tone of smooth muscle strips was measured by isometric force transducers. Length of smooth muscle cells (SMC) isolated from the IAS in the basal state was determined by phase contrast microscopy. Experiments were repeated in the presence of Y 27632, a ROCK inhibitor. Involvement of the RhoA/ROCK machinery was analyzed by reverse-transcription polymerase chain reaction, Western blot, and immunocytochemistry. Reversal of H-ras knock-out effect was evaluated by transfection of SMCs with the constitutively activated (G12V) mutant. RESULTS Basal tone of the H-ras(-/-) IAS was significantly higher and resistant to relaxation by Y 27632, compared with the H-ras(+/+) IAS. Similarly, the length of SMCs from H-ras(-/-) IAS was significantly shorter. Y 27632 eliminated this difference. RhoA immunoreactivity shifted from cytoplasm to plasma membrane in H-ras(-/-) SMCs, a change typically associated with contraction. Further, SMCs from H-ras(-/-) mice exhibited higher levels of the contractile proteins ROCK II, phosphorylated-MYPT(1) and phosphorylated-MLC(20). Transfection with the G12V mutant increased the length of H-ras(-/-) cells. Conversely, the dominant negative H-ras (S17N) mutant decreased the length of H-ras(+/+) cells. CONCLUSIONS H-ras negatively regulates basal tone in the IAS by inhibiting RhoA/Rho-kinase machinery. Studies may have significant relevance in the pathophysiology and therapy of certain anorectal motility disorders associated with the IAS dysfunction.
Collapse
Affiliation(s)
- Márcio A F de Godoy
- Departments of Medicine, Division of Gastroenterology & Hepatology, Pharmacology and Experimental Therapeutics, and Emergency Medicine, Jefferson Medical College of Thomas Jefferson University, Philadelphia, Pennyslvania 19107, USA
| | | | | | | | | | | |
Collapse
|
42
|
Ball SG, Shuttleworth CA, Kielty CM. Platelet-derived growth factor receptor-α is a key determinant of smooth muscle α-actin filaments in bone marrow-derived mesenchymal stem cells. Int J Biochem Cell Biol 2007; 39:379-91. [PMID: 17070723 DOI: 10.1016/j.biocel.2006.09.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2006] [Revised: 09/04/2006] [Accepted: 09/05/2006] [Indexed: 11/22/2022]
Abstract
Smooth muscle alpha-actin filaments are a defining feature of mesenchymal stem cells, and of mesenchyme-derived contractile smooth muscle cells, pericytes and myofibroblasts. Here, we show that adult bone marrow-derived mesenchymal stem cells express abundant cell surface platelet-derived growth factor receptor-alpha, having a high ratio to platelet-derived growth factor receptor-beta. Signaling through platelet-derived growth factor receptor-alpha increases smooth muscle alpha-actin filaments by activating RhoA, which results in Rho-associated kinase (ROCK)-dependent cofilin phosphorylation, enhancing smooth muscle alpha-actin filament polymerization, and also upregulates smooth muscle alpha-actin expression. In contrast, platelet-derived growth factor receptor-beta signaling strongly upregulates RhoE, which inhibits ROCK activity, promoting smooth muscle alpha-actin filament depolymerization. This study thus provides new insights into the distinct roles of platelet-derived growth factor receptor-alpha and -beta signaling in regulating the adult mesenchymal stem cell contractile cytoskeleton.
Collapse
Affiliation(s)
- Stephen G Ball
- UK Centre for Tissue Engineering, Faculty of Life Sciences, The University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | | | | |
Collapse
|
43
|
Mueller L, Goumas FA, Himpel S, Brilloff S, Rogiers X, Broering DC. Imatinib mesylate inhibits proliferation and modulates cytokine expression of human cancer-associated stromal fibroblasts from colorectal metastases. Cancer Lett 2006; 250:329-38. [PMID: 17141949 DOI: 10.1016/j.canlet.2006.10.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2006] [Revised: 10/17/2006] [Accepted: 10/23/2006] [Indexed: 01/26/2023]
Abstract
Recent data have expanded the concept that cancer-associated stromal fibroblasts (CAFs) play an important role in tumor invasion and angiogenesis. Here, we show that platelet-derived growth factor (PDGF) is a mitogen for human CAFs isolated from hepatic metastases of colorectal cancer. The tyrosine kinase inhibitor imatinib mesylate (1 microM) abrogated the PDGF-induced DNA synthesis, and furthermore counteracted an inhibitory effect of PDGF on the expression of alpha-smooth muscle actin (alpha-SMA). High-dose imatinib mesylate (10 microM) decreased the viability of CAFs in vitro independent from co-stimulation with PDGF. Interestingly, imatinib mesylate (10 microM) strikingly induced the expression of the pro-inflammatory and pro-angiogenic cytokines interleukin (IL)-6 and IL-8, and mildly stimulated the release of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF). Our results suggest that imatinib mesylate, due to its anti-proliferative activity, may be effective in combination with other substances for the treatment of colorectal metastasis progression.
Collapse
Affiliation(s)
- Lars Mueller
- Department of Hepatobiliary Surgery and Solid Organ Transplantation, University Hospital Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | | | | | | | | | | |
Collapse
|
44
|
Miano JM, Long X, Fujiwara K. Serum response factor: master regulator of the actin cytoskeleton and contractile apparatus. Am J Physiol Cell Physiol 2006; 292:C70-81. [PMID: 16928770 DOI: 10.1152/ajpcell.00386.2006] [Citation(s) in RCA: 371] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Serum response factor (SRF) is a highly conserved and widely expressed, single copy transcription factor that theoretically binds up to 1,216 permutations of a 10-base pair cis element known as the CArG box. SRF-binding sites were defined initially in growth-related genes. Gene inactivation or knockdown studies in species ranging from unicellular eukaryotes to mice have consistently shown loss of SRF to be incompatible with life. However, rather than being critical for proliferation and growth, these genetic studies point to a crucial role for SRF in cellular migration and normal actin cytoskeleton and contractile biology. In fact, recent genomic studies reveal nearly half of the >200 SRF target genes encoding proteins with functions related to actin dynamics, lamellipodial/filopodial formation, integrin-cytoskeletal coupling, myofibrillogenesis, and muscle contraction. SRF has therefore emerged as a dispensable transcription factor for cellular growth but an absolutely essential orchestrator of actin cytoskeleton and contractile homeostasis. This review summarizes the recent genomic and genetic analyses of CArG-SRF that support its role as an ancient, master regulator of the actin cytoskeleton and contractile machinery.
Collapse
Affiliation(s)
- Joseph M Miano
- Cardiovascular Research Institute, University of Rochester School of Medicine, 601 Elmwood Avenue, Rochester, NY 14642, USA.
| | | | | |
Collapse
|
45
|
Risinger GM, Hunt TS, Updike DL, Bullen EC, Howard EW. Matrix metalloproteinase-2 expression by vascular smooth muscle cells is mediated by both stimulatory and inhibitory signals in response to growth factors. J Biol Chem 2006; 281:25915-25. [PMID: 16854986 DOI: 10.1074/jbc.m513513200] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In response to growth factors, vascular smooth muscle cells (VSMCs) undergo a phenotypic modulation from a contractile, non-proliferative state to an activated, migratory state. This transition is characterized by changes in their gene expression profile, particularly by a significant down-regulation of contractile proteins. Platelet-derived growth factor (PDGF)-BB has long been known to initiate VSMC de-differentiation and mitogenesis. Insulin-like growth factor (IGF)-I, on the other hand, has differing effects depending on the model studied. Here, we report that both IGF-I and PDGF-BB stimulated VSMC de-differentiation of rat heart-derived SMCs in culture, although only PDGF-BB was capable of inducing proliferation. Although both PDGF-BB and IGF-I stimulation resulted in decreased smooth muscle alpha-actin expression and increased matrix metalloproteinase (MMP)-2 expression, the response to IGF-I was significantly more rapid. The increased MMP-2 expression in response to both growth factors was due to increased transcription rates and was dependent on the action of phosphatidylinositol 3-kinase (PI3K) and its downstream effector, Akt. Both PDGF-BB and IGF-I activated PI3K/Akt to similar degrees; however, only PDGF-BB concomitantly stimulated an inhibitory signaling pathway that antagonized the effects of Akt but did not alter the extent or duration of Akt activation. Together, these findings suggest that changes in MMP-2 expression are part of the program of VSMC phenotypic modulation and that both PDGF-BB and IGF-I, despite their different abilities to induce proliferation in this model, are capable of inducing VSMC activation.
Collapse
Affiliation(s)
- George M Risinger
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | | | | | | | | |
Collapse
|
46
|
Garat CV, Fankell D, Erickson PF, Reusch JEB, Bauer NN, McMurtry IF, Klemm DJ. Platelet-derived growth factor BB induces nuclear export and proteasomal degradation of CREB via phosphatidylinositol 3-kinase/Akt signaling in pulmonary artery smooth muscle cells. Mol Cell Biol 2006; 26:4934-48. [PMID: 16782881 PMCID: PMC1489168 DOI: 10.1128/mcb.02477-05] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2005] [Revised: 02/01/2006] [Accepted: 04/14/2006] [Indexed: 01/05/2023] Open
Abstract
Cyclic AMP response element binding protein (CREB) content is diminished in smooth muscle cells (SMCs) in remodeled pulmonary arteries from animals with pulmonary hypertension and in the SMC layers of atherogenic systemic arteries and cardiomyocytes from hypertensive individuals. Loss of CREB can be induced in cultured SMCs by chronic exposure to hypoxia or platelet-derived growth factor BB (PDGF-BB). Here we investigated the signaling pathways and mechanisms by which PDGF elicits depletion of SMC CREB. Chronic PDGF treatment increased CREB ubiquitination in SMCs, while treatment of SMCs with the proteasome inhibitor lactacystin prevented decreases in CREB content. The nuclear export inhibitor leptomycin B also prevented depletion of SMC CREB alone or in combination with lactacystin. Subsequent studies showed that PDGF activated extracellular signal-regulated kinase, Jun N-terminal protein kinase, and phosphatidylinositol 3 (PI3)-kinase pathways in SMCs. Inhibition of these pathways blocked SMC proliferation in response to PDGF, but only inhibition of PI3-kinase or its effector, Akt, blocked PDGF-induced CREB loss. Finally, chimeric proteins containing enhanced cyan fluorescent protein linked to wild-type CREB or CREB molecules with mutations in several recognized phosphorylation sites were introduced into SMCs. PDGF treatment reduced the levels of each of these chimeric proteins except for one containing mutations in adjacent serine residues (serines 103 and 107), suggesting that CREB loss was dependent on CREB phosphorylation at these sites. We conclude that PDGF stimulates nuclear export and proteasomal degradation of CREB in SMCs via PI3-kinase/Akt signaling. These results indicate that in addition to direct phosphorylation, proteolysis and intracellular localization are key mechanisms regulating CREB content and activity in SMCs.
Collapse
Affiliation(s)
- Chrystelle V Garat
- Cardiovascular Pulmonary Research, University of Colorado Health Sciences Center, 4200 East Ninth Ave., Campus Box B-133, Denver, CO 80262, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Han M, Wen JK, Zheng B, Cheng Y, Zhang C. Serum deprivation results in redifferentiation of human umbilical vascular smooth muscle cells. Am J Physiol Cell Physiol 2006; 291:C50-8. [PMID: 16467401 DOI: 10.1152/ajpcell.00524.2005] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Phenotypic change of vascular smooth muscle cells (VSMCs) from a differentiated to a dedifferentiated state accompanies the early stage of atherosclerosis and restenosis. Although much progress has been made in determining the molecular mechanisms involved in VSMC dedifferentiation, research on VSMC redifferentiation is hindered by the lack of an appropriate complete redifferentiation model. We established an in vitro model of redifferentiation by using postconfluent VSMCs from human umbilical artery. We demonstrated that serum-deprived VSMCs are capable of complete redifferentiation. After serum deprivation, postconfluent cultured human umbilical VSMCs became elongated and spindle shaped, with elevation of myofilament density, and reacquired contraction. Expressions of VSMC-specific contractile proteins, such as smooth muscle (SM) alpha-actin, SM-myosin heavy chain, calponin, and SM 22alpha, were increased and reached the levels in differentiated cells after serum deprivation. To determine the molecular mechanism of the phenotypic reversion, the levels of expression, phosphorylation, and binding activity of serum response factor (SRF), a key phenotypic modulator for VSMCs, were measured. The results showed that SRF binding activity with CArG motif was significantly increased after serum deprivation, whereas no changes were found in SRF expression and phosphorylation. The increased SRF binding activity was accompanied by an increase in expression of its coactivators such as myocardin. Furthermore, the phenotypic reversion was markedly inhibited by decoy double-strand oligodeoxynucleotides containing SM alpha-actin CArG motif, which was able to competitively bind to SRF. The results suggested that serum deprivation results in redifferentiation of human umbilical VSMCs. This novel model of VSMC phenotypic reversion should be valuable for research on vascular disease.
Collapse
Affiliation(s)
- Mei Han
- Department of Biochemistry and Molecular Biology, Hebei Medical University, No. 361, Zhongshan East Road, Shijiazhuang, 050017, China
| | | | | | | | | |
Collapse
|
48
|
Romano F, Chiarenza C, Palombi F, Filippini A, Padula F, Ziparo E, De Cesaris P. Platelet-derived growth factor-BB-induced hypertrophy of peritubular smooth muscle cells is mediated by activation of p38 MAP-kinase and of Rho-kinase. J Cell Physiol 2006; 207:123-31. [PMID: 16270352 DOI: 10.1002/jcp.20554] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Peritubular smooth muscle cells (PSMC) from rat testis in primary serum-free cultures unexpectedly undergo contraction and subsequent cell hypertrophy in response to the growth factor PDGF-BB, remaining stationary. The present study investigates the transduction pathways involved in the observed paradoxical upregulation of the differentiated phenotype and induction of hypertrophy in PSMC. PI3K, ERK, JNK, and p38 kinases, known to mediate PDGF-BB signaling in the canonic dedifferentiative and proliferative response of smooth muscle cells (SMC) were rapidly activated by PDGF-BB but only p38 remained activated after 2-day stimulation. Immunofluorescence and immunoblotting experiments showed that in 4-day treatment: (i) continuous inhibition of PI3K, of ERK, of JNK, failed to inhibit either cell enlargement and formation of prominent alpha-SM actin containing stress fibers or the typical increase in alpha-SM actin; (ii) when stimulated in the presence of the p38 inhibitor SB203580 both responses were significantly inhibited and cytofluorimetric analysis of cell size showed a remarkable reduction of the hypertrophic response. PDGF-BB was also found to activate the small GTPase RhoA and inhibition of Rho-dependent kinase ROCK by Y27632 counteracted the effects of PDGF-BB similarly to SB203580. Both the transcription factor ATF2 and the nucleosomal kinase MSK1, downstream targets of p38, were activated by PDGF-BB, but p38 inhibitor SB203580 inhibited only the phosphorylation of MSK1 which appeared unaffected by ROCK inhibitor Y27632. In concluding, p38 and the Rho-ROCK system were found to play prominent, probably independent roles in the upregulation of PSMC differentiated phenotype and induction of hypertrophy by PDGF-BB.
Collapse
Affiliation(s)
- Francesca Romano
- Istituto Pasteur, Fondazione Cenci Bolognetti, Department of Histology and Medical Embryology, La Sapienza University of Rome, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
49
|
Hamada K, Sasaki T, Koni PA, Natsui M, Kishimoto H, Sasaki J, Yajima N, Horie Y, Hasegawa G, Naito M, Miyazaki JI, Suda T, Itoh H, Nakao K, Mak TW, Nakano T, Suzuki A. The PTEN/PI3K pathway governs normal vascular development and tumor angiogenesis. Genes Dev 2005; 19:2054-65. [PMID: 16107612 PMCID: PMC1199575 DOI: 10.1101/gad.1308805] [Citation(s) in RCA: 234] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PTEN is an important tumor suppressor gene. Hereditary mutation of PTEN causes tumor-susceptibility diseases such as Cowden disease. We used the Cre-loxP system to generate an endothelial cell-specific mutation of Pten (Tie2CrePten) in mice. Tie2CrePten(flox/+) mice displayed enhanced tumorigenesis due to an increase in angiogenesis driven by vascular growth factors. This effect was partially dependent on the PI3K subunits p85alpha and p110gamma. In vitro, Tie2CrePten(flox/+) endothelial cells showed enhanced proliferation/migration. Tie2CrePten(flox/flox) mice died before embryonic day 11.5 (E11.5) due to bleeding and cardiac failure caused by impaired recruitment of pericytes and vascular smooth muscle cells to blood vessels, and of cardiomyocytes to the endocardium. These phenotypes depend strongly on p110gamma rather than on p85alpha and were associated with decreased expression of Ang-1, VCAM-1, connexin 40, and ephrinB2 but increased expression of Ang-2, VEGF-A, VEGFR1, and VEGFR2. Pten is thus indispensable for normal cardiovascular morphogenesis and post-natal angiogenesis, including tumor angiogenesis.
Collapse
MESH Headings
- Animals
- Base Sequence
- Cells, Cultured
- Female
- Fetal Heart/embryology
- Fetal Heart/metabolism
- Genes, Tumor Suppressor
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Mutant Strains
- Mice, Transgenic
- Mutation
- Neoplasms, Experimental/blood supply
- Neoplasms, Experimental/genetics
- Neovascularization, Pathologic
- Neovascularization, Physiologic
- PTEN Phosphohydrolase
- Phosphatidylinositol 3-Kinases/deficiency
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/physiology
- Protein Tyrosine Phosphatases/deficiency
- Protein Tyrosine Phosphatases/genetics
- Protein Tyrosine Phosphatases/physiology
- RNA, Small Interfering/genetics
- Tumor Suppressor Proteins/deficiency
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/physiology
Collapse
Affiliation(s)
- Koichi Hamada
- Department of Molecular Biology, Department of Microbiology, Department of Gastroenterology, Akita University School of Medicine, Akita 010-8543, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Kaplan-Albuquerque N, Van Putten V, Weiser-Evans MC, Nemenoff RA. Depletion of serum response factor by RNA interference mimics the mitogenic effects of platelet derived growth factor-BB in vascular smooth muscle cells. Circ Res 2005; 97:427-33. [PMID: 16081871 DOI: 10.1161/01.res.0000179776.40216.a9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Promoters of many smooth muscle-specific genes (SM-genes) contain multiple CArG boxes, which represent a binding site for serum response factor (SRF). Transcriptional control through these regions involves interactions with SRF and specific coactivators such as myocardin. We have previously reported that suppression of SM-gene expression by platelet derived growth factor (PDGF) is associated with redistribution of SRF, leading to lower intra-nuclear levels, and a reduction in SRF transactivation. To further assess the role of SRF depletion on VSMC phenotype, the current study used RNA interference (RNAi). Two SRF-specific sequences constructed as hairpins were stably expressed in rat VSMC. Clones expressing SRF RNAi had no detectable SRF expression by immunoblotting, and showed diminished levels of SM alpha-actin protein and promoter activity. Unexpectedly, depletion of VSMC resulted in increased rates of proliferation and migration. Several genes whose expression is increased by PDGF stimulation, including c-Jun, were similarly induced in cells lacking SRF. Effects of SRF depletion were not attributable to altered PDGF receptor activity or alterations in activation of Akt. These data indicate that loss of SRF transactivation in VSMC, in this case through suppression via RNAi, induces biological responses similar to that seen with PDGF.
Collapse
|