1
|
Patterson SD, Massett ME, Huang X, Jørgensen HG, Michie AM. The MYC-NFATC2 axis maintains the cell cycle and mitochondrial function in acute myeloid leukaemia cells. Mol Oncol 2024; 18:2234-2254. [PMID: 38459421 PMCID: PMC11467801 DOI: 10.1002/1878-0261.13630] [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: 11/16/2023] [Revised: 01/30/2024] [Accepted: 02/29/2024] [Indexed: 03/10/2024] Open
Abstract
Acute myeloid leukaemia (AML) is a clonal haematological malignancy affecting the myeloid lineage, with generally poor patient outcomes owing to the lack of targeted therapies. The histone lysine demethylase 4A (KDM4A) has been established as a novel therapeutic target in AML, due to its selective oncogenic role within leukaemic cells. We identify that the transcription factor nuclear factor of activated T cells 2 (NFATC2) is a novel binding and transcriptional target of KDM4A in the human AML THP-1 cell line. Furthermore, cytogenetically diverse AML cell lines, including THP-1, were dependent on NFATC2 for colony formation in vitro, highlighting a putative novel mechanism of AML oncogenesis. Our study demonstrates that NFATC2 maintenance of cell cycle progression in human AML cells was driven primarily by CCND1. Through RNA sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq), NFATc2 was shown to bind to the promoter region of genes involved in oxidative phosphorylation and subsequently regulate their gene expression in THP-1 cells. Furthermore, our data show that NFATC2 shares transcriptional targets with the transcription factor c-MYC, with MYC knockdown phenocopying NFATC2 knockdown. These data suggest a newly identified co-ordinated role for NFATC2 and MYC in the maintenance of THP-1 cell function, indicative of a potential means of therapeutic targeting in human AML.
Collapse
Affiliation(s)
- Shaun D. Patterson
- Paul O'Gorman Leukaemia Research Centre, Gartnavel General HospitalUniversity of GlasgowUK
| | - Matthew E. Massett
- Paul O'Gorman Leukaemia Research Centre, Gartnavel General HospitalUniversity of GlasgowUK
| | - Xu Huang
- Paul O'Gorman Leukaemia Research Centre, Gartnavel General HospitalUniversity of GlasgowUK
| | - Heather G. Jørgensen
- Paul O'Gorman Leukaemia Research Centre, Gartnavel General HospitalUniversity of GlasgowUK
| | - Alison M. Michie
- Paul O'Gorman Leukaemia Research Centre, Gartnavel General HospitalUniversity of GlasgowUK
| |
Collapse
|
2
|
Hanaki S, Habara M, Tomiyasu H, Sato Y, Miki Y, Masaki T, Shibutani S, Shimada M. NFAT activation by FKBP52 promotes cancer cell proliferation by suppressing p53. Life Sci Alliance 2024; 7:e202302426. [PMID: 38803221 PMCID: PMC11109481 DOI: 10.26508/lsa.202302426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
FK506-binding protein 52 (FKBP52) is a member of the FKBP family of proline isomerases. FKBP52 is up-regulated in various cancers and functions as a positive regulator of steroid hormone receptors. Depletion of FKBP52 is known to inhibit cell proliferation; however, the detailed mechanism remains poorly understood. In this study, we found that FKBP52 depletion decreased MDM2 transcription, leading to stabilization of p53, and suppressed cell proliferation. We identified NFATc1 and NFATc3 as transcription factors that regulate MDM2 We also found that FKBP52 associated with NFATc3 and facilitated its nuclear translocation. In addition, calcineurin, a well-known Ca2+ phosphatase essential for activation of NFAT, plays a role in MDM2 transcription. Supporting this notion, MDM2 expression was found to be regulated by intracellular Ca2+ Taken together, these findings reveal a new role of FKBP52 in promoting cell proliferation via the NFAT-MDM2-p53 axis, and indicate that inhibition of FKBP52 could be a new therapeutic tool to activate p53 and inhibit cell proliferation.
Collapse
Affiliation(s)
- Shunsuke Hanaki
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi, Japan
| | - Makoto Habara
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi, Japan
| | - Haruki Tomiyasu
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi, Japan
| | - Yuki Sato
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi, Japan
| | - Yosei Miki
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi, Japan
| | - Takahiro Masaki
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi, Japan
| | - Shusaku Shibutani
- Department of Veterinary Hygiene, Yamaguchi University, Yamaguchi, Japan
| | - Midori Shimada
- Department of Veterinary Biochemistry, Yamaguchi University, Yamaguchi, Japan
- Department of Molecular Biology, Nagoya University, Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
3
|
Vanderhout RJ, Abdalla EA, Leishman EM, Barbut S, Wood BJ, Baes CF. Genetic architecture of white striping in turkeys (Meleagris gallopavo). Sci Rep 2024; 14:9007. [PMID: 38637585 PMCID: PMC11026500 DOI: 10.1038/s41598-024-59309-8] [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: 11/17/2023] [Accepted: 04/09/2024] [Indexed: 04/20/2024] Open
Abstract
White striping (WS) is a myopathy of growing concern to the turkey industry. It is rising in prevalence and has negative consequences for consumer acceptance and the functional properties of turkey meat. The objective of this study was to conduct a genome-wide association study (GWAS) and functional analysis on WS severity. Phenotypic data consisted of white striping scored on turkey breast fillets (N = 8422) by trained observers on a 0-3 scale (none to severe). Of the phenotyped birds, 4667 genotypic records were available using a proprietary 65 K single nucleotide polymorphism (SNP) chip. The SNP effects were estimated using a linear mixed model with a 30-SNP sliding window approach used to express the percentage genetic variance explained. Positional candidate genes were those located within 50 kb of the top 1% of SNP windows explaining the most genetic variance. Of the 95 positional candidate genes, seven were further classified as functional candidate genes because of their association with both a significant gene ontology and molecular function term. The results of the GWAS emphasize the polygenic nature of the trait with no specific genomic region contributing a large portion to the overall genetic variance. Significant pathways relating to growth, muscle development, collagen formation, circulatory system development, cell response to stimulus, and cytokine production were identified. These results help to support published biological associations between WS and hypoxia and oxidative stress and provide information that may be useful for future-omics studies in understanding the biological associations with WS development in turkeys.
Collapse
Affiliation(s)
- Ryley J Vanderhout
- Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
- Hybrid Turkeys, 650 Riverbend Drive Suite C, Kitchener, ON, N2K 3S2, Canada
| | - Emhimad A Abdalla
- Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
- Vereinigte Informationssysteme Tierhaltung W.V. (Vit), Heinrich-Schröder-Weg 1, 27283, Verden, Germany
| | - Emily M Leishman
- Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Shai Barbut
- Department of Food Science, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Benjamin J Wood
- Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
- Hybrid Turkeys, 650 Riverbend Drive Suite C, Kitchener, ON, N2K 3S2, Canada
- School of Veterinary Science, University of Queensland, Gatton, QLD, 4343, Australia
| | - Christine F Baes
- Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada.
- Institute of Genetics, Vetsuisse Faculty, University of Bern, 3001, Bern, Switzerland.
| |
Collapse
|
4
|
Dupuy M, Gueguinou M, Potier-Cartereau M, Lézot F, Papin M, Chantôme A, Rédini F, Vandier C, Verrecchia F. SK Ca- and Kv1-type potassium channels and cancer: Promising therapeutic targets? Biochem Pharmacol 2023; 216:115774. [PMID: 37678626 DOI: 10.1016/j.bcp.2023.115774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023]
Abstract
Ion channels are transmembrane structures that allow the passage of ions across cell membranes such as the plasma membrane or the membranes of various organelles like the nucleus, endoplasmic reticulum, Golgi apparatus or mitochondria. Aberrant expression of various ion channels has been demonstrated in several tumor cells, leading to the promotion of key functions in tumor development, such as cell proliferation, resistance to apoptosis, angiogenesis, invasion and metastasis. The link between ion channels and these key biological functions that promote tumor development has led to the classification of cancers as oncochannelopathies. Among all ion channels, the most varied and numerous, forming the largest family, are the potassium channels, with over 70 genes encoding them in humans. In this context, this review will provide a non-exhaustive overview of the role of plasma membrane potassium channels in cancer, describing 1) the nomenclature and structure of potassium channels, 2) the role of these channels in the control of biological functions that promotes tumor development such as proliferation, migration and cell death, and 3) the role of two particular classes of potassium channels, the SKCa- and Kv1- type potassium channels in cancer progression.
Collapse
Affiliation(s)
- Maryne Dupuy
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000 Nantes, France.
| | | | | | - Frédéric Lézot
- Sorbonne University, INSERM UMR933, Hôpital Trousseau (AP-HP), Paris F-75012, France
| | - Marion Papin
- N2C UMR 1069, University of Tours, INSERM, Tours, France
| | | | - Françoise Rédini
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000 Nantes, France
| | | | - Franck Verrecchia
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000 Nantes, France.
| |
Collapse
|
5
|
Canonical Wnt Pathway Is Involved in Chemoresistance and Cell Cycle Arrest Induction in Colon Cancer Cell Line Spheroids. Int J Mol Sci 2023; 24:ijms24065252. [PMID: 36982333 PMCID: PMC10049556 DOI: 10.3390/ijms24065252] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 03/12/2023] Open
Abstract
The presence of cancer stem cells (CSCs) has been associated with the induction of drug resistance and disease recurrence after therapy. 5-Fluorouracil (5FU) is widely used as the first-line treatment of colorectal cancer (CRC). However, its effectiveness may be limited by the induction of drug resistance in tumor cells. The Wnt pathway plays a key role in the development and CRC progression, but it is not clearly established how it is involved in CSCs resistance to treatment. This work aimed to investigate the role played by the canonical Wnt/β-catenin pathway in CSCs resistance to 5FU treatment. Using tumor spheroids as a model of CSCs enrichment of CRC cell lines with different Wnt/β-catenin contexts, we found that 5FU induces in all CRC spheroids tested cell death, DNA damage, and quiescence, but in different proportions for each one: RKO spheroids were very sensitive to 5FU, while SW480 were less susceptible, and the SW620 spheroids, the metastatic derivative of SW480 cells, displayed the highest resistance to death, high clonogenic capacity, and the highest ability for regrowth after 5FU treatment. Activating the canonical Wnt pathway with Wnt3a in RKO spheroids decreased the 5FU-induced cell death. But the Wnt/β-catenin pathway inhibition with Adavivint alone or in combination with 5FU in spheroids with aberrant activation of this pathway produced a severe cytostatic effect compromising their clonogenic capacity and diminishing the stem cell markers expression. Remarkably, this combined treatment also induced the survival of a small cell subpopulation that could exit the arrest, recover SOX2 levels, and re-grow after treatment.
Collapse
|
6
|
KBTBD11, encoding a novel PPARγ target gene, is involved in NFATc1 proteolysis by interacting with HSC70 and HSP60. Sci Rep 2022; 12:20273. [PMID: 36434268 PMCID: PMC9700792 DOI: 10.1038/s41598-022-24929-5] [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: 06/13/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022] Open
Abstract
We previously revealed that Kbtbd11 mRNA levels increase during 3T3-L1 differentiation and Kbtbd11 knockdown suppresses whereas its overexpression promotes adipogenesis. However, how Kbtbd11 mRNA is regulated during adipocyte differentiation and how the KBTBD11 protein functions in adipocytes remain elusive. This study aimed to examine the transcriptional regulatory mechanism of Kbtbd11 during adipocyte differentiation, KBTBD11-interacting protein functions, and elucidate the role of KBTBD11 in adipocytes. First, we identified the PPRE consensus sequences in the Kbtbd11 exon 1- and intron 1-containing region and demonstrated that PPARγ acts on this region to regulate Kbtbd11 expression. Next, we purified the KBTBD11 protein complex from 3T3-L1 adipocytes and identified heat shock proteins HSC70 and HSP60 as novel KBTBD11-interacting proteins. HSC70 and HSP60 inhibition increased KBTBD11 protein levels that promoted NFATc1 ubiquitination. These data suggest that HSC70 and HSP60 are involved in KBTBD11 stabilization and are responsible for NFATc1 regulation on the protein level. In summary, this study describes first the protein regulatory mechanism of NFATc1 through the HSC70/HSP60-KBTBD11 interaction that could provide a potential new target for the differentiation and proliferation of various cells, including adipocytes and tumors.
Collapse
|
7
|
Huynh DTN, Jin Y, Van Nguyen D, Myung CS, Heo KS. Ginsenoside Rh1 Inhibits Angiotensin II-Induced Vascular Smooth Muscle Cell Migration and Proliferation through Suppression of the ROS-Mediated ERK1/2/p90RSK/KLF4 Signaling Pathway. Antioxidants (Basel) 2022; 11:antiox11040643. [PMID: 35453328 PMCID: PMC9030830 DOI: 10.3390/antiox11040643] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 02/01/2023] Open
Abstract
Vascular smooth muscle cell (VSMC) proliferation and migration play key roles in the progression of atherosclerosis and restenosis. A variety of ginsenosides exert various cardiovascular benefits. However, whether and how ginsenoside Rh1 (Rh1) inhibits VSMC dysfunction remain unclear. Here, we investigated the inhibitory effects of Rh1 on rat aortic smooth muscle cell (RASMC) migration and proliferation induced by angiotensin II (Ang II) and the underlying mechanisms. Cell proliferation and migration were evaluated using sulforhodamine B and wound-healing assay. The molecular mechanisms were investigated using Western blotting, quantitative reverse-transcription polymerase chain reaction analysis, immunofluorescence staining, and luciferase assay. Reactive oxygen species (ROS) production was measured using dihydroethidium and MitoSOX staining. We found that Rh1 dose-dependently suppressed Ang II-induced cell proliferation and migration. Concomitantly, Ang II increased protein levels of osteopontin, vimentin, MMP2, MMP9, PCNA, and cyclin D1, while these were reduced by Rh1 pretreatment. Notably, Ang II enhanced both the protein expression and promoter activity of KLF4, a key regulator of phenotypic switching, whereas pretreatment with Rh1 reversed these effects. Mechanistically, the effects of Rh1 on VSMC proliferation and migration were found to be associated with inhibition of ERK1/2/p90RSK signaling. Furthermore, the inhibitory effects of Rh1 were accompanied by inhibition of ROS production. In conclusion, Rh1 inhibited the Ang II-induced migration and proliferation of RASMCs by suppressing the ROS-mediated ERK1/2/p90RSK signaling pathway.
Collapse
Affiliation(s)
- Diem Thi Ngoc Huynh
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, 99 Daehak-ro, Yuseong-Gu, Daejeon 34134, Korea; (D.T.N.H.); (Y.J.); (D.V.N.); (C.-S.M.)
- Department of Pharmacy, Da Nang University of Medical Technology and Pharmacy, Da Nang 550000, Vietnam
| | - Yujin Jin
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, 99 Daehak-ro, Yuseong-Gu, Daejeon 34134, Korea; (D.T.N.H.); (Y.J.); (D.V.N.); (C.-S.M.)
| | - Dung Van Nguyen
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, 99 Daehak-ro, Yuseong-Gu, Daejeon 34134, Korea; (D.T.N.H.); (Y.J.); (D.V.N.); (C.-S.M.)
| | - Chang-Seon Myung
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, 99 Daehak-ro, Yuseong-Gu, Daejeon 34134, Korea; (D.T.N.H.); (Y.J.); (D.V.N.); (C.-S.M.)
| | - Kyung-Sun Heo
- College of Pharmacy and Institute of Drug Research and Development, Chungnam National University, 99 Daehak-ro, Yuseong-Gu, Daejeon 34134, Korea; (D.T.N.H.); (Y.J.); (D.V.N.); (C.-S.M.)
- Correspondence: ; Tel.: +82-42-821-5927
| |
Collapse
|
8
|
Decoding the Phosphatase Code: Regulation of Cell Proliferation by Calcineurin. Int J Mol Sci 2022; 23:ijms23031122. [PMID: 35163061 PMCID: PMC8835043 DOI: 10.3390/ijms23031122] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 02/06/2023] Open
Abstract
Calcineurin, a calcium-dependent serine/threonine phosphatase, integrates the alterations in intracellular calcium levels into downstream signaling pathways by regulating the phosphorylation states of several targets. Intracellular Ca2+ is essential for normal cellular physiology and cell cycle progression at certain critical stages of the cell cycle. Recently, it was reported that calcineurin is activated in a variety of cancers. Given that abnormalities in calcineurin signaling can lead to malignant growth and cancer, the calcineurin signaling pathway could be a potential target for cancer treatment. For example, NFAT, a typical substrate of calcineurin, activates the genes that promote cell proliferation. Furthermore, cyclin D1 and estrogen receptors are dephosphorylated and stabilized by calcineurin, leading to cell proliferation. In this review, we focus on the cell proliferative functions and regulatory mechanisms of calcineurin and summarize the various substrates of calcineurin. We also describe recent advances regarding dysregulation of the calcineurin activity in cancer cells. We hope that this review will provide new insights into the potential role of calcineurin in cancer development.
Collapse
|
9
|
Kai W, Lin C, Jin Y, Ping-Lin H, Xun L, Bastian A, Arnulf S, Sha-Sha X, Xu L, Shu C. Urethral meatus stricture BOO stimulates bladder smooth muscle cell proliferation and pyroptosis via IL‑1β and the SGK1‑NFAT2 signaling pathway. Mol Med Rep 2020; 22:219-226. [PMID: 32468047 PMCID: PMC7248470 DOI: 10.3892/mmr.2020.11092] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 03/25/2020] [Indexed: 11/29/2022] Open
Abstract
Bladder outlet obstruction (BOO), which is primarily caused by benign prostatic hyperplasia, is a common chronic disease. However, previous studies have most commonly investigated BOO using the acute obstruction model. In the present study, a chronic obstruction model was established to investigate the different pathological alterations in the bladder between acute and chronic obstruction. Compared with chronic obstruction, acute obstruction led to increased expression of proliferating cell nuclear antigen and interleukin-1β, which are markers of proliferation and inflammation, respectively. Furthermore, increased fibrosis in the bladder at week 2 was observed. Low pressure promoted mice bladder smooth muscle cell (MBSMC) proliferation, and pressure overload inhibited cell proliferation and increased the proportion of dead MBSMCs. Further investigation using serum/glucocorticoid regulated kinase 1 (SGK1) small interfering RNAs indicated that low pressure may promote MBSMC proliferation by upregulating SGK1 and nuclear factor of activated T-cell expression levels. Therefore, the present study suggested that acute obstruction led to faster decompensation of bladder function and chronic bladder obstruction displayed an enhanced ability to progress to BOO.
Collapse
Affiliation(s)
- Wang Kai
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610041, P.R. China
| | - Chen Lin
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610041, P.R. China
| | - Yang Jin
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610041, P.R. China
| | - He Ping-Lin
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610041, P.R. China
| | - Liu Xun
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610041, P.R. China
| | - Amend Bastian
- Department of Urology, University of Tübingen, D‑72070 Tübingen, Baden‑Württemberg, Germany
| | - Stenzl Arnulf
- Department of Urology, University of Tübingen, D‑72070 Tübingen, Baden‑Württemberg, Germany
| | - Xing Sha-Sha
- Central Laboratory, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Luo Xu
- Department of Urology, Zunyi Medical University, Guiyang, Guizhou 563000, P.R. China
| | - Cui Shu
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
10
|
Feng S, Gao L, Zhang D, Tian X, Kong L, Shi H, Wu L, Huang Z, Du B, Liang C, Zhang Y, Yao R. MiR-93 regulates vascular smooth muscle cell proliferation, and neointimal formation through targeting Mfn2. Int J Biol Sci 2019; 15:2615-2626. [PMID: 31754334 PMCID: PMC6854371 DOI: 10.7150/ijbs.36995] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 07/28/2019] [Indexed: 11/05/2022] Open
Abstract
Background/Aims: Vascular smooth muscle cell (VSMC) hyperplasia plays important roles in the pathogenesis of many vascular diseases, such as atherosclerosis and restenosis. Many microRNAs (miRs) have recently been reported to regulate the proliferation and migration of VSMC. In the current study, we aimed to explore the function of miR-93 in VSMCs and its molecular mechanism. Methods: First, qRT-PCR and immunofluorescence assays were performed to determine miR-93 expression in rat VSMCs following carotid artery injury in vivo and platelet-derived growth factor-BB (PDGF-BB) stimulation in vitro. Next, the biological role of miR-93 in rat VSMC proliferation and migration was examined in vivo and vitro. EdU incorporation assay and MTT assay for measuring cell proliferation, Transwell cell invasion assay and Cell scratch wound assay for measuring cell migration. Then, the targets of miR-93 were identified. Finally, the expression levels of proteins in the Raf-ERK1/2 pathway were measured by western blot. Results: MiR-93 was upregulated in rat VSMCs following carotid artery injury in vivo. Similar results were observed in ex vivo cultured VSMCs after PDGF-BB treatment. MiR-93 inhibition suppressed neointimal formation after carotid artery injury. Moreover, our results demonstrated that a miR-93 inhibitor suppressed the PDGF-BB induced proliferation and migration of in VSMC. This inhibitor also decreased the expression levels of MMP2 and cyclin D1. Mechanistically, we discovered that mitofusin 2(Mfn2) is a direct target of miR-93. Furthermore, an analysis of the signaling events revealed that miR-93-mediated VSMC proliferation and migration occurred via the Raf-ERK1/2 pathway. Conclusions: Our findings suggest that miR-93 promotes VSMCs proliferation and migration by targeting Mfn2. MiR-93 may be a new target for treating in-stent restenosis.
Collapse
Affiliation(s)
- Shengdong Feng
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lu Gao
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dianhong Zhang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinyu Tian
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lingyao Kong
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huiting Shi
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Leiming Wu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhen Huang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Binbin Du
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Cui Liang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanzhou Zhang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rui Yao
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Goshima T, Habara M, Maeda K, Hanaki S, Kato Y, Shimada M. Calcineurin regulates cyclin D1 stability through dephosphorylation at T286. Sci Rep 2019; 9:12779. [PMID: 31484966 PMCID: PMC6726757 DOI: 10.1038/s41598-019-48976-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 08/16/2019] [Indexed: 12/27/2022] Open
Abstract
The Calcineurin/NFAT (nuclear factor of activated T cells) pathway plays an essential role in the tumorigenic and metastatic properties in breast cancer. The molecular mechanism of the antiproliferative effect of calcineurin inhibition, however, is poorly understood. We found that calcineurin inhibition delayed cell cycle progression at G1/S, and promoted cyclin D1 degradation by inhibiting dephosphorylation at T286. Importantly, overexpression of cyclin D1 partially rescued delayed G1/S progression, thereby revealing cyclin D1 as a key factor downstream of calcineurin inhibition. Cyclin D1 upregulation is observed in human invasive breast cancers, and our findings indicate that dysregulation of T286 phosphorylation could play a role in this phenomenon. We therefore propose that targeting site specific phosphorylation of cyclin D1 could be a potential strategy for clinical intervention of invasive breast cancer.
Collapse
Affiliation(s)
- Takahiro Goshima
- Department of Cell Biology, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Makoto Habara
- Department of Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Keisuke Maeda
- Department of Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Shunsuke Hanaki
- Department of Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Yoichi Kato
- Department of Cell Biology, Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Midori Shimada
- Department of Biochemistry, Joint Faculty of Veterinary Science, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan.
| |
Collapse
|
12
|
Govatati S, Pichavaram P, Janjanam J, Zhang B, Singh NK, Mani AM, Traylor JG, Orr AW, Rao GN. NFATc1-E2F1-LMCD1-Mediated IL-33 Expression by Thrombin Is Required for Injury-Induced Neointima Formation. Arterioscler Thromb Vasc Biol 2019; 39:1212-1226. [PMID: 31043075 PMCID: PMC6540998 DOI: 10.1161/atvbaha.119.312729] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective- IL (interleukin)-33 has been shown to play a role in endothelial dysfunction, but its role in atherosclerosis is controversial. Therefore, the purpose of this study is to examine its role in vascular wall remodeling following injury. Approach and Results- Thrombin induced IL-33 expression in a time-dependent manner in human aortic smooth muscle cells and inhibition of its activity by its neutralizing antibody suppressed thrombin induced human aortic smooth muscle cell migration but not DNA synthesis. In exploring the mechanisms, we found that Par1 (protease-activated receptor 1), Gαq/11 (Gα protein q/11), PLCβ3 (phospholipase Cβ3), NFATc1 (nuclear factor of activated T cells), E2F1 (E2F transcription factor 1), and LMCD1 (LIM and cysteine-rich domains protein 1) are involved in thrombin-induced IL-33 expression and migration. Furthermore, we identified an NFAT-binding site at -100 nt that mediates thrombin-induced IL-33 promoter activity. Interestingly, we observed that NFATc1, E2F1, and LMCD1 bind to NFAT site in response to thrombin and found that LMCD1, while alone has no significant effect, enhanced either NFATc1 or E2F1-dependent IL-33 promoter activity. In addition, we found that guidewire injury induces IL-33 expression in SMC and its neutralizing antibodies substantially reduce SMC migration and neointimal growth in vivo. Increased expression of IL-33 was also observed in human atherosclerotic lesions as compared to arteries without any lesions. Conclusions- The above findings reveal for the first time that thrombin-induced human aortic smooth muscle cell migration and injury-induced neointimal growth require IL-33 expression. In addition, thrombin-induced IL-33 expression requires LMCD1 enhanced combinatorial activation of NFATc1 and E2F1.
Collapse
MESH Headings
- Animals
- Binding Sites
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Co-Repressor Proteins/genetics
- Co-Repressor Proteins/metabolism
- Disease Models, Animal
- E2F1 Transcription Factor/genetics
- E2F1 Transcription Factor/metabolism
- Female
- Femoral Artery/drug effects
- Femoral Artery/injuries
- Femoral Artery/metabolism
- Femoral Artery/pathology
- HEK293 Cells
- Humans
- Interleukin-33/genetics
- Interleukin-33/metabolism
- LIM Domain Proteins/genetics
- LIM Domain Proteins/metabolism
- Male
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- NFATC Transcription Factors/genetics
- NFATC Transcription Factors/metabolism
- Neointima
- Promoter Regions, Genetic
- Signal Transduction
- Up-Regulation
- Vascular System Injuries/genetics
- Vascular System Injuries/metabolism
- Vascular System Injuries/pathology
Collapse
Affiliation(s)
- Suresh Govatati
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Prahalathan Pichavaram
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jagadeesh Janjanam
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Baolin Zhang
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Nikhlesh K. Singh
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Arul M. Mani
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - James G. Traylor
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA
| | - A. Wayne Orr
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA
| | - Gadiparthi N. Rao
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
13
|
Ding N, Geng B, Li Z, Yang Q, Yan L, Wan L, Zhang B, Wang C, Xia Y. Fluid shear stress promotes osteoblast proliferation through the NFATc1-ERK5 pathway. Connect Tissue Res 2019; 60:107-116. [PMID: 29609502 DOI: 10.1080/03008207.2018.1459588] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Extracellular-regulated kinase 5 (ERK5) is thought to regulate osteoblast proliferation. To further understand how ERK5 signaling regulates osteoblast proliferation induced by fluid shear stress (FSS), we examined some potential signaling targets associated with ERK5 in MC3T3-E1 cells. METHODS MC3T3-E1 cells were treated with XMD8-92 (an ERK5 inhibitor) or Cyclosporin A (CsA, a nuclear factor of activated T cells (NFAT) c1 inhibitor) and/or exposed to 12 dyn/cm2 FSS. Phosphorylated-ERK5 (p-ERK5) and expression levels of NFATc1, ERK5, E2F2, and cyclin E1 were analyzed by western blot. The mRNA levels of genes associated with cell proliferation were analyzed by Polymerase Chain Reaction (PCR) array. Subcellular localization of p-ERK5 and NFATc1 were determined by immunofluorescence. Cell proliferation was evaluated by MTT assay. RESULTS NFATc1 expression was up-regulated by FSS. XMD8-92 only blocked ERK5 activation; however, CsA decreased NFATc1 and p-ERK5 levels, including after FSS stimulation. Exposure to NFATc1 inhibitor or ERK5 inhibitor resulted in decreased E2F2 and cyclin E1 expression and proliferation by proliferative MC3T3-E1 cells. Furthermore, immunofluorescence results illustrated that NFATc1 induced ERK5 phosphorylation, resulting in p-ERK5 translocation to the nucleus. CONCLUSIONS Our results reveal that NFATc1 acts as an intermediate to promote the phosphorylation of ERK5 induced by FSS. Moreover, activated NFATc1-ERK5 signaling up-regulates the expression of E2F2 and cyclin E1, which promote osteoblast proliferation.
Collapse
Affiliation(s)
- Ning Ding
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Bin Geng
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Zhonghao Li
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Quanzeng Yang
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Liang Yan
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Lang Wan
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Bo Zhang
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Cuifang Wang
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| | - Yayi Xia
- a Department of Orthopaedics , Lanzhou University Second Hospital , Lanzhou , Gansu , China.,b Orthopaedics Key Laboratory of Gansu Province , Lanzhou , Gansu , China
| |
Collapse
|
14
|
Sompol P, Norris CM. Ca 2+, Astrocyte Activation and Calcineurin/NFAT Signaling in Age-Related Neurodegenerative Diseases. Front Aging Neurosci 2018; 10:199. [PMID: 30038565 PMCID: PMC6046440 DOI: 10.3389/fnagi.2018.00199] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022] Open
Abstract
Mounting evidence supports a fundamental role for Ca2+ dysregulation in astrocyte activation. Though the activated astrocyte phenotype is complex, cell-type targeting approaches have revealed a number of detrimental roles of activated astrocytes involving neuroinflammation, release of synaptotoxic factors and loss of glutamate regulation. Work from our lab and others has suggested that the Ca2+/calmodulin dependent protein phosphatase, calcineurin (CN), provides a critical link between Ca2+ dysregulation and the activated astrocyte phenotype. A proteolyzed, hyperactivated form of CN appears at high levels in activated astrocytes in both human tissue and rodent tissue around regions of amyloid and vascular pathology. Similar upregulation of the CN-dependent transcription factor nuclear factor of activated T cells (NFAT4) also appears in activated astrocytes in mouse models of Alzheimer's disease (ADs) and traumatic brain injury (TBI). Major consequences of hyperactivated CN/NFAT4 signaling in astrocytes are neuroinflammation, synapse dysfunction and glutamate dysregulation/excitotoxicity, which will be covered in this review article.
Collapse
Affiliation(s)
- Pradoldej Sompol
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Christopher M Norris
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, United States.,Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| |
Collapse
|
15
|
Singh NK, Janjanam J, Rao GN. p115 RhoGEF activates the Rac1 GTPase signaling cascade in MCP1 chemokine-induced vascular smooth muscle cell migration and proliferation. J Biol Chem 2017; 292:14080-14091. [PMID: 28655771 DOI: 10.1074/jbc.m117.777896] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 06/20/2017] [Indexed: 12/30/2022] Open
Abstract
Although the involvement of Rho proteins in the pathogenesis of vascular diseases is well studied, little is known about the role of their upstream regulators, the Rho guanine nucleotide exchange factors (RhoGEFs). Here, we sought to identify the RhoGEFs involved in monocyte chemotactic protein 1 (MCP1)-induced vascular wall remodeling. We found that, among the RhoGEFs tested, MCP1 induced tyrosine phosphorylation of p115 RhoGEF but not of PDZ RhoGEF or leukemia-associated RhoGEF in human aortic smooth muscle cells (HASMCs). Moreover, p115 RhoGEF inhibition suppressed MCP1-induced HASMC migration and proliferation. Consistent with these observations, balloon injury (BI) induced p115 RhoGEF tyrosine phosphorylation in rat common carotid arteries, and siRNA-mediated down-regulation of its levels substantially attenuated BI-induced smooth muscle cell migration and proliferation, resulting in reduced neointima formation. Furthermore, depletion of p115 RhoGEF levels also abrogated MCP1- or BI-induced Rac1-NFATc1-cyclin D1-CDK6-PKN1-CDK4-PAK1 signaling, which, as we reported previously, is involved in vascular wall remodeling. Our findings also show that protein kinase N1 (PKN1) downstream of Rac1-cyclin D1/CDK6 and upstream of CDK4-PAK1 in the p115 RhoGEF-Rac1-NFATc1-cyclin D1-CDK6-PKN1-CDK4-PAK1 signaling axis is involved in the modulation of vascular wall remodeling. Of note, we also observed that CCR2-Gi/o-Fyn signaling mediates MCP1-induced p115 RhoGEF and Rac1 GTPase activation. These findings suggest that p115 RhoGEF is critical for MCP1-induced HASMC migration and proliferation in vitro and for injury-induced neointima formation in vivo by modulating Rac1-NFATc1-cyclin D1-CDK6-PKN1-CDK4-PAK1 signaling.
Collapse
Affiliation(s)
- Nikhlesh K Singh
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163.
| | - Jagadeesh Janjanam
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Gadiparthi N Rao
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163.
| |
Collapse
|
16
|
|
17
|
Tan J, Yang L, Liu C, Yan Z. MicroRNA-26a targets MAPK6 to inhibit smooth muscle cell proliferation and vein graft neointimal hyperplasia. Sci Rep 2017; 7:46602. [PMID: 28429763 PMCID: PMC5399463 DOI: 10.1038/srep46602] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 03/21/2017] [Indexed: 12/21/2022] Open
Abstract
Neointima formation is the major reason for vein graft failure. However, the underlying mechanism is unclear. The aim of this study was to determine the role of miR-26a in the development of neointimal hyperplasia of autogenous vein grafts. Using autologous jugular vein grafts in the rat carotid artery as a model, we found that miR-26a was significantly downregulated in grafted veins as well as proliferating vascular smooth muscle cells (VSMCs) stimulated with platelet-derived growth factor-BB (PDGF-BB). Overexpression of miR-26a reduced the proliferation and migration of VSMCs. Further analysis revealed that the effects of miR-26a in VSMCs were mediated by targeting MAPK6 at the mRNA and protein levels. Luciferase assays showed that miR-26a repressed wild type (WT) MAPK6-3′-UTR-luciferase activity but not mutant MAPK6-3′-UTR-luciferease reporter. MAPK6 deficiency reduced proliferation and migration; in contrast, overexpression of MAPK6 enhanced the proliferation and migration of VSMCs. This study confirmed that neointimal hyperplasia in vein grafts was reduced in vivo by up-regulated miR-26a expression. In conclusion, our results showed that miR-26a is an important regulator of VSMC functions and neointimal hyperplasia, suggesting that miR-26a may be a potential therapeutic target for autologous vein graft diseases.
Collapse
Affiliation(s)
- Juanjuan Tan
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Liguo Yang
- Department of Cardiology, Shanghai Jiao Tong University afliated Sixth People's Hospital South Campus, Shanghai, 201400, P. R. China
| | - Cuicui Liu
- Central laboratory, Shanghai Jiao Tong University afliated Sixth People's Hospital South Campus, Shanghai, 201400, P. R. China
| | - Zhiqiang Yan
- Central laboratory, Shanghai Jiao Tong University afliated Sixth People's Hospital South Campus, Shanghai, 201400, P. R. China
| |
Collapse
|
18
|
Cheng W, Yan K, Xie LY, Chen F, Yu HC, Huang YX, Dang CX. MiR-143-3p controls TGF-β1-induced cell proliferation and extracellular matrix production in airway smooth muscle via negative regulation of the nuclear factor of activated T cells 1. Mol Immunol 2016; 78:133-139. [PMID: 27639060 DOI: 10.1016/j.molimm.2016.09.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/05/2016] [Accepted: 09/06/2016] [Indexed: 12/22/2022]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that function in diverse biological processes. However, little is known about the precise role of microRNAs in the functioning of airway smooth muscle cells (ASMCs). Here, we investigated the potential role and mechanisms of the miR-143 -3p on proliferation and the extracellular matrix (ECM) protein production of ASMCs. We demonstrated that miR-143-3p was aberrantly lower in ASMCs isolated from individuals with asthma than in individuals without asthma. Meanwhile, TGF-β1 caused a marked decrease in a time-dependent manner in miR-143-3p expression in ASMCs from asthmatics. Additionally, the overexpression of miR- 143-3p robustly reduced TGF-β1-induced ASMCs proliferation and downregulated CDK and cyclin expression, whereas the inhibition of miR-143-3p significantly enhanced ASMCs proliferation and upregulated the level of CDKs and cyclins. Re-expression of miR-143-3p attenuated ECM protein deposition reflected as a marked decrease in the expression of type I collagen and fibronectin, whereas miR-143-3p downregulation caused an opposite effect on the expression of type I collagen and fibronectin. Moreover, qRT-PCR and western blot analysis indicated that miR-143-3p negatively regulated the expression of nuclear factor of activated T cells 1 (NFATc1). Subsequent analyses demonstrated that NFATc1 was a direct and functional target of miR-143-3p, which was validated by the dual luciferase reporter assay. Most importantly, the overexpression of NFATc1 effectively reversed the inhibition of miR-143-3p on TGF-β1-induced proliferation, and strikingly abrogated the effect of miR-143-3p on the expression of CDK4 and Cyclin D1. Together, miR-143-3p may function as an inhibitor of asthma airway remodeling by suppressing proliferation and ECM protein deposition in TGF-β1-mediated ASMCs via the negative regulation of NFATc1 signaling, suggesting miR-143-3p as a potential therapeutic target for asthma.
Collapse
Affiliation(s)
- Wei Cheng
- The Second Department of Respiratory Internal Medicine, Xi'an Children's Hospital, Xi'an 710003, Shaanxi, PR China; Department of Oncology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, PR China.
| | - Kun Yan
- Department of General Surgery, the Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710004, Shaanxi, PR China
| | - Li-Yi Xie
- Department of Nephropathy, the First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, PR China.
| | - Feng Chen
- Department of Internal Medicine, Xi'an Children's Hospital, Xi'an 710003,Shaanxi, PR China
| | - Hong-Chuan Yu
- Department of Internal Medicine, Xi'an Children's Hospital, Xi'an 710003,Shaanxi, PR China
| | - Yan-Xia Huang
- Department of Internal Medicine, Xi'an Children's Hospital, Xi'an 710003,Shaanxi, PR China
| | - Cheng-Xue Dang
- Department of Oncology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, PR China
| |
Collapse
|
19
|
Cell cycle and apoptosis regulation by NFAT transcription factors: new roles for an old player. Cell Death Dis 2016; 7:e2199. [PMID: 27100893 PMCID: PMC4855676 DOI: 10.1038/cddis.2016.97] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/13/2016] [Accepted: 03/16/2016] [Indexed: 12/11/2022]
Abstract
The NFAT (nuclear factor of activated T cells) family of transcription factors consists of four Ca2+-regulated members (NFAT1–NFAT4), which were first described in T lymphocytes. In addition to their well-documented role in T lymphocytes, where they control gene expression during cell activation and differentiation, NFAT proteins are also expressed in a wide range of cells and tissue types and regulate genes involved in cell cycle, apoptosis, angiogenesis and metastasis. The NFAT proteins share a highly conserved DNA-binding domain (DBD), which allows all NFAT members to bind to the same DNA sequence in enhancers or promoter regions. The same DNA-binding specificity suggests redundant roles for the NFAT proteins, which is true during the regulation of some genes such as IL-2 and p21. However, it has become increasingly clear that different NFAT proteins and even isoforms can have unique functions. In this review, we address the possible reasons for these distinct roles, particularly regarding N- and C-terminal transactivation regions (TADs) and the partner proteins that interact with these TADs. We also discuss the genes regulated by NFAT during cell cycle regulation and apoptosis and the role of NFAT during tumorigenesis.
Collapse
|
20
|
Fric J, Lim CXF, Mertes A, Lee BTK, Viganò E, Chen J, Zolezzi F, Poidinger M, Larbi A, Strobl H, Zelante T, Ricciardi-Castagnoli P. Calcium and calcineurin-NFAT signaling regulate granulocyte-monocyte progenitor cell cycle via Flt3-L. Stem Cells 2015; 32:3232-44. [PMID: 25100642 PMCID: PMC4282522 DOI: 10.1002/stem.1813] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 07/07/2014] [Indexed: 12/11/2022]
Abstract
Maintenance of myeloid progenitor cells is controlled by complex regulatory mechanisms and is orchestrated by multiple different transcription factors. Here, we report that the activation of the transcription factor nuclear factor of activated T cells (NFAT) by calcium-sensing protein calcineurin inhibits the proliferation of myeloid granulocyte–monocyte progenitors (GMPs). Myeloid progenitor subtypes exhibit variable sensitivity to induced Ca2+ entry and consequently display differential engagement of the calcineurin-NFAT pathway. This study shows that inhibition of the calcineurin-NFAT pathway enhances the proliferation of GMPs both in vitro and in vivo and demonstrates that calcineurin-NFAT signaling in GMPs is initiated by Flt3-L. Inhibition of the calcineurin-NFAT pathway modified expression of the cell cycle regulation genes Cdk4, Cdk6, and Cdkn1a (p21), thus enabling rapid cell cycle progression specifically in GMPs. NFAT inhibitor drugs are extensively used in the clinic to restrict the pathological activation of lymphoid cells, and our data reveal for the first time that these therapies also exert potent effects on maintenance of the myeloid cell compartment through specific regulation of GMP proliferation. Stem Cells2014;32:3232–3244
Collapse
Affiliation(s)
- Jan Fric
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Xu F, Ahmed ASI, Kang X, Hu G, Liu F, Zhang W, Zhou J. MicroRNA-15b/16 Attenuates Vascular Neointima Formation by Promoting the Contractile Phenotype of Vascular Smooth Muscle Through Targeting YAP. Arterioscler Thromb Vasc Biol 2015; 35:2145-52. [PMID: 26293467 DOI: 10.1161/atvbaha.115.305748] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/10/2015] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To investigate the functional role of the microRNA (miR)-15b/16 in vascular smooth muscle (SM) phenotypic modulation. APPROACH AND RESULTS We found that miR-15b/16 is one of the most abundant mRs expressed in contractile vascular smooth muscle cells (VSMCs). However, when contractile VSMCs get converted to a synthetic phenotype, miR-15b/16 expression is significantly reduced. Knocking down endogenous miR-15b/16 in VSMCs attenuates SM-specific gene expression but promotes VSMC proliferation and migration. Conversely, overexpression of miR-15b/16 promotes SM contractile gene expression while attenuating VSMC migration and proliferation. Consistent with this, overexpression of miR-15b/16 in a rat carotid balloon injury model markedly attenuates injury-induced SM dedifferentiation and neointima formation. Mechanistically, we identified the potent oncoprotein yes-associated protein (YAP) as a downstream target of miR-15b/16 in VSMCs. Reporter assays validated that miR-15b/16 targets YAP's 3' untranslated region. Moreover, overexpression of miR-15b/16 significantly represses YAP expression, whereas conversely, depletion of endogenous miR-15b/16 results in upregulation of YAP expression. CONCLUSIONS These results indicate that miR-15b/16 plays a critical role in SM phenotypic modulation at least partly through targeting YAP. Restoring expression of miR-15b/16 would be a potential therapeutic approach for treatment of proliferative vascular diseases.
Collapse
Affiliation(s)
- Fei Xu
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.)
| | - Abu Shufian Ishtiaq Ahmed
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.)
| | - Xiuhua Kang
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.)
| | - Guoqing Hu
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.)
| | - Fang Liu
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.)
| | - Wei Zhang
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.)
| | - Jiliang Zhou
- From the Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China (F.X., X.K., W.Z.); and Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta (A.S.I.A., X.K., G.H., F.L., J.Z.).
| |
Collapse
|
22
|
Chin-Smith EC, Willey FR, Slater DM, Taggart MJ, Tribe RM. Nuclear factor of activated T-cell isoform expression and regulation in human myometrium. Reprod Biol Endocrinol 2015; 13:83. [PMID: 26238508 PMCID: PMC4523953 DOI: 10.1186/s12958-015-0086-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 07/30/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND During pregnancy, myometrial gene and protein expression is tightly regulated to accommodate fetal growth, promote quiescence and ultimately prepare for the onset of labour. It is proposed that changes in calcium signalling, may contribute to regulating gene expression and that nuclear factor of activated T-cell (NFAT) transcription factors (isoforms c1-c4) may be involved. Currently, there is little information regarding NFAT expression and regulation in myometrium. METHODS This study examined NFAT isoform mRNA expression in human myometrial tissue and cells from pregnant women using quantitative PCR. The effects of the Ca(2+) ionophore A23187 and in vitro stretch (25 % elongation, static strain; Flexercell FX-4000 Tension System) on NFAT expression were determined in cultured human myometrial cells. RESULTS Human myometrial tissue and cultured cells expressed NFATc1-c4 mRNA. NFATc2 gene expression in cultured cells was increased in response to 6 h stretch (11.5 fold, P < 0.001, n = 6) and calcium ionophore (A23187, 5 μM) treatment (20.6 fold, P < 0.001, n = 6). This response to stretch was significantly reduced (90 %, P < 0.001, n = 10) in the presence of an intracellular calcium chelator, BAPTA-AM (20 μM). CONCLUSIONS These data suggest that NFATc2 expression is regulated by intracellular calcium and in vitro stretch, and that the stretch response in human myometrial cells is dependent upon intracellular calcium signalling pathways. Our findings indicate a potentially unique role for NFATc2 in mediating stretch-induced gene expression per se and warrant further exploration in relation to the mechanisms promoting uterine smooth muscle growth in early pregnancy and/or labour.
Collapse
Affiliation(s)
- Evonne C Chin-Smith
- Division of Women's Health, King's College London, Women's Health Academic Centre KHP, St Thomas' Hospital, 10th Floor, North Wing, Westminster Bridge Road, London, SE1 7EH, UK.
| | - Frances R Willey
- Division of Women's Health, King's College London, Women's Health Academic Centre KHP, St Thomas' Hospital, 10th Floor, North Wing, Westminster Bridge Road, London, SE1 7EH, UK.
| | - Donna M Slater
- Physiology and Pharmacology, Cumming School of Medicine, Alberta Children's Hospital Research Institute for Child and Maternal Health, University of Calgary, Alberta, T2N 4 N1, Canada.
| | - Michael J Taggart
- Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK.
| | - Rachel M Tribe
- Division of Women's Health, King's College London, Women's Health Academic Centre KHP, St Thomas' Hospital, 10th Floor, North Wing, Westminster Bridge Road, London, SE1 7EH, UK.
| |
Collapse
|
23
|
Kim MH, Ham O, Lee SY, Choi E, Lee CY, Park JH, Lee J, Seo HH, Seung M, Choi E, Min PK, Hwang KC. MicroRNA-365 inhibits the proliferation of vascular smooth muscle cells by targeting cyclin D1. J Cell Biochem 2015; 115:1752-61. [PMID: 24819721 DOI: 10.1002/jcb.24841] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 05/08/2014] [Indexed: 02/02/2023]
Abstract
Abnormal proliferation of vascular smooth muscle cells (VSMCs) is a common feature of disease progression in atherosclerosis. Cell proliferation is regulated by cell cycle regulatory proteins. MicroRNAs (miR) have been reported to act as important gene regulators and play essential roles in the proliferation and migration of VSMCs in a cardiovascular disease. However, the roles and mechanisms of miRs in VSMCs and neointimal formation are far from being fully understood. In this study, cell cycle-specific cyclin D1 was found to be a potential target of miR-365 by direct binding. Through an in vitro experiment, we showed that exogenous miR-365 overexpression reduced VSMC proliferation and proliferating cell nuclear antigen (PCNA) expression, while miR-365 was observed to block G1/S transition in platelet-derived growth factor-bb (PDGF-bb)-induced VSMCs. In addition, the proliferation of VSMCs by various stimuli, including PDGF-bb, angiotensin II (Ang II), and serum, led to the downregulation of miR-365 expression levels. The expression of miR-365 was confirmed in balloon-injured carotid arteries. Taken together, our results suggest an anti-proliferative role for miR-365 in VSMC proliferation, at least partly via modulating the expression of cyclin D1. Therefore, miR-365 may influence neointimal formation in atherosclerosis patients.
Collapse
Affiliation(s)
- Myung-Hyun Kim
- Cardiology Division, Heart Center, Gangnam Severance Hospital, Yonsei University College of Medicine, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Holtz BJ, Lodewyk KB, Sebolt-Leopold JS, Ernst SA, Williams JA. ERK activation is required for CCK-mediated pancreatic adaptive growth in mice. Am J Physiol Gastrointest Liver Physiol 2014; 307:G700-10. [PMID: 25104499 PMCID: PMC4187068 DOI: 10.1152/ajpgi.00163.2014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
High levels of cholecystokinin (CCK) can stimulate pancreatic adaptive growth in which mature acinar cells divide, leading to enhanced pancreatic mass with parallel increases in protein, DNA, RNA, and digestive enzyme content. Prolonged release of CCK can be induced by feeding trypsin inhibitor (TI) to disrupt normal feedback control. This leads to exocrine growth in a CCK-dependent manner. The extracellular signal-related kinase (ERK) pathway regulates many proliferative processes in various tissues and disease models. The aim of this study was to evaluate the role of ERK signaling in pancreatic adaptive growth using the MEK inhibitors PD-0325901 and trametinib (GSK-1120212). It was determined that PD-0325901 given two times daily by gavage or mixed into powdered chow was an effective and specific inhibitor of ERK signaling in vivo. TI-containing chow led to a robust increase in pancreatic mass, protein, DNA, and RNA content. This pancreatic adaptive growth was blocked in mice fed chow containing the MEK inhibitors. PD-0325901 blocked TI-induced ERK-regulated early response genes, cell-cycle proteins, and mitogenesis by acinar cells. It was determined that ERK signaling is necessary for the initiation of pancreatic adaptive growth but not necessary to maintain it. PD-0325901 blocked adaptive growth when given before cell-cycle initiation but not after mitogenesis had been established. Furthermore, GSK-1120212, a chemically distinct inhibitor of the ERK pathway that is now approved for clinical use, inhibited growth similar to PD-0325901. These data demonstrate that the ERK pathway is required for CCK-stimulated pancreatic adaptive growth.
Collapse
Affiliation(s)
- Bryan J. Holtz
- 1Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan;
| | - Kevin B. Lodewyk
- 1Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan;
| | | | - Stephen A. Ernst
- 3Department of Cellular and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan; and
| | - John A. Williams
- 1Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan; ,4Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
25
|
Store-operated CRAC channels regulate gene expression and proliferation in neural progenitor cells. J Neurosci 2014; 34:9107-23. [PMID: 24990931 DOI: 10.1523/jneurosci.0263-14.2014] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Calcium signals regulate many critical processes during vertebrate brain development including neurogenesis, neurotransmitter specification, and axonal outgrowth. However, the identity of the ion channels mediating Ca(2+) signaling in the developing nervous system is not well defined. Here, we report that embryonic and adult mouse neural stem/progenitor cells (NSCs/NPCs) exhibit store-operated Ca(2+) entry (SOCE) mediated by Ca(2+) release-activated Ca(2+) (CRAC) channels. SOCE in NPCs was blocked by the CRAC channel inhibitors La(3+), BTP2, and 2-APB and Western blots revealed the presence of the canonical CRAC channel proteins STIM1 and Orai1. Knock down of STIM1 or Orai1 significantly diminished SOCE in NPCs, and SOCE was lost in NPCs from transgenic mice lacking Orai1 or STIM1 and in knock-in mice expressing the loss-of-function Orai1 mutant, R93W. Therefore, STIM1 and Orai1 make essential contributions to SOCE in NPCs. SOCE in NPCs was activated by epidermal growth factor and acetylcholine, the latter occurring through muscarinic receptors. Activation of SOCE stimulated gene transcription through calcineurin/NFAT (nuclear factor of activated T cells) signaling through a mechanism consistent with local Ca(2+) signaling by Ca(2+) microdomains near CRAC channels. Importantly, suppression or deletion of STIM1 and Orai1 expression significantly attenuated proliferation of embryonic and adult NPCs cultured as neurospheres and, in vivo, in the subventricular zone of adult mice. These findings show that CRAC channels serve as a major route of Ca(2+) entry in NPCs and regulate key effector functions including gene expression and proliferation, indicating that CRAC channels are important regulators of mammalian neurogenesis.
Collapse
|
26
|
Afroze T, Yang G, Khoshbin A, Tanwir M, Tabish T, Momen A, Husain M. Calcium efflux activity of plasma membrane Ca2+ ATPase-4 (PMCA4) mediates cell cycle progression in vascular smooth muscle cells. J Biol Chem 2014; 289:7221-7231. [PMID: 24448801 DOI: 10.1074/jbc.m113.533638] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We explored the role played by plasma membrane calcium ATPase-4 (PMCA4) and its alternative splice variants in the cell cycle of vascular smooth muscle cells (VSMC). A novel variant (PMCA4e) was discovered. Quantitative real-time-PCR-quantified PMCA4 splice variant proportions differed in specific organs. The PMCA4a:4b ratio in uninjured carotid arteries (∼1:1) was significantly reduced by wire denudation injury (to ∼1:3) by modulation of alternative splicing, as confirmed by novel antibodies against PMCA4a/e and PMCA4b. Laser capture microdissection localized this shift to the media and adventitia. Primary carotid VSMC from PMCA4 knock-out (P4KO) mice showed impaired [(3)H]thymidine incorporation and G1 phase arrest as compared with wild type (P4WT). Electroporation of expression constructs encoding PMCA4a, PMCA4b, and a PMCA4b mutant lacking PDZ binding rescued this phenotype of P4KO cells, whereas a mutant with only 10% of normal Ca(2+) efflux activity could not. Microarray of early G1-synchronized VSMC showed 39-fold higher Rgs16 (NFAT (nuclear factor of activated T-cells) target; MAPK inhibitor) and 69-fold higher Decorin (G1 arrest marker) expression in P4KO versus P4WT. Validation by Western blot also revealed decreased levels of Cyclin D1 and NFATc3 in P4KO. Microarrays of P4KO VSMC rescued by PMCA4a or PMCA4b expression showed reversal of perturbed Rgs16, Decorin, and NFATc3 expression levels. However, PMCA4a rescue caused a 44-fold reduction in AP-2β, a known anti-proliferative transcription factor, whereas PMCA4b rescue resulted in a 50-fold reduction in p15 (Cyclin D1/Cdk4 inhibitor). We conclude that Ca(2+) efflux activity of PMCA4 underlies G1 progression in VSMC and that PMCA4a and PMCA4b differentially regulate specific downstream mediators.
Collapse
Affiliation(s)
- Talat Afroze
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Ge Yang
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Amir Khoshbin
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Mansoor Tanwir
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Taha Tabish
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Abdul Momen
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Mansoor Husain
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada; Institute of Medical Sciences, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Ontario M5G 1L7; Department of Medicine, University of Toronto, Toronto, Ontario M5G 2C4, Canada.
| |
Collapse
|
27
|
Kundumani-Sridharan V, Singh NK, Kumar S, Gadepalli R, Rao GN. Nuclear factor of activated T cells c1 mediates p21-activated kinase 1 activation in the modulation of chemokine-induced human aortic smooth muscle cell F-actin stress fiber formation, migration, and proliferation and injury-induced vascular wall remodeling. J Biol Chem 2013; 288:22150-62. [PMID: 23737530 DOI: 10.1074/jbc.m113.454082] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Recent literature suggests that cyclin-dependent kinases (CDKs) mediate cell migration. However, the mechanisms were not known. Therefore, the objective of this study is to test whether cyclin/CDKs activate Pak1, an effector of Rac1, whose involvement in the modulation of cell migration and proliferation is well established. Monocyte chemotactic protein 1 (MCP1) induced Pak1 phosphorylation/activation in human aortic smooth muscle cells (HASMCs) in a delayed time-dependent manner. MCP1 also stimulated F-actin stress fiber formation in a delayed manner in HASMCs, as well as the migration and proliferation of these cells. Inhibition of Pak1 suppressed MCP1-induced HASMC F-actin stress fiber formation, migration, and proliferation. MCP1 induced cyclin D1 expression as well as CDK6 and CDK4 activities, and these effects were dependent on activation of NFATc1. Depletion of NFATc1, cyclin D1, CDK6, or CDK4 levels attenuated MCP1-induced Pak1 phosphorylation/activation and resulted in decreased HASMC F-actin stress fiber formation, migration, and proliferation. CDK4, which appeared to be activated downstream of CDK6, formed a complex with Pak1 in response to MCP1. MCP1 also activated Rac1 in a time-dependent manner, and depletion/inhibition of its levels/activation abrogated MCP1-induced NFATc1-cyclin D1-CDK6-CDK4-Pak1 signaling and, thereby, decreased HASMC F-actin stress fiber formation, migration, and proliferation. In addition, smooth muscle-specific deletion of NFATc1 led to decreased cyclin D1 expression and CDK6, CDK4, and Pak1 activities, resulting in reduced neointima formation in response to injury. Thus, these observations reveal that Pak1 is a downstream effector of CDK4 and Rac1-dependent, NFATc1-mediated cyclin D1 expression and CDK6 activity mediate this effect. In addition, smooth muscle-specific deletion of NFATc1 prevented the capacity of vascular smooth muscle cells for MCP-1-induced activation of the cyclin D1-CDK6-CDK4-Pak1 signaling axis, affecting their migration and proliferation in vitro and injury-induced neointima formation in vivo.
Collapse
|
28
|
Zetterqvist AV, Berglund LM, Blanco F, Garcia-Vaz E, Wigren M, Dunér P, Andersson AMD, To F, Spegel P, Nilsson J, Bengtsson E, Gomez MF. Inhibition of nuclear factor of activated T-cells (NFAT) suppresses accelerated atherosclerosis in diabetic mice. PLoS One 2013; 8:e65020. [PMID: 23755169 PMCID: PMC3670844 DOI: 10.1371/journal.pone.0065020] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 04/21/2013] [Indexed: 01/13/2023] Open
Abstract
Objective of the Study Diabetic patients have a much more widespread and aggressive form of atherosclerosis and therefore, higher risk for myocardial infarction, peripheral vascular disease and stroke, but the molecular mechanisms leading to accelerated damage are still unclear. Recently, we showed that hyperglycemia activates the transcription factor NFAT in the arterial wall, inducing the expression of the pro-atherosclerotic protein osteopontin. Here we investigate whether NFAT activation may be a link between diabetes and atherogenesis. Methodology and Principal Findings Streptozotocin (STZ)-induced diabetes in apolipoprotein E−/− mice resulted in 2.2 fold increased aortic atherosclerosis and enhanced pro-inflammatory burden, as evidenced by elevated blood monocytes, endothelial activation- and inflammatory markers in aorta, and pro-inflammatory cytokines in plasma. In vivo treatment with the NFAT blocker A-285222 for 4 weeks completely inhibited the diabetes-induced aggravation of atherosclerosis, having no effect in non-diabetic mice. STZ-treated mice exhibited hyperglycemia and higher plasma cholesterol and triglycerides, but these were unaffected by A-285222. NFAT-dependent transcriptional activity was examined in aorta, spleen, thymus, brain, heart, liver and kidney, but only augmented in the aorta of diabetic mice. A-285222 completely blocked this diabetes-driven NFAT activation, but had no impact on the other organs or on splenocyte proliferation or cytokine secretion, ruling out systemic immunosuppression as the mechanism behind reduced atherosclerosis. Instead, NFAT inhibition effectively reduced IL-6, osteopontin, monocyte chemotactic protein 1, intercellular adhesion molecule 1, CD68 and tissue factor expression in the arterial wall and lowered plasma IL-6 in diabetic mice. Conclusions Targeting NFAT signaling may be a novel and attractive approach for the treatment of diabetic macrovascular complications.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Apolipoproteins E/deficiency
- Apolipoproteins E/metabolism
- Atherosclerosis/blood
- Atherosclerosis/complications
- Atherosclerosis/pathology
- Biomarkers/metabolism
- Blood Glucose/metabolism
- Body Weight/drug effects
- Cholesterol/blood
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Disease Progression
- Inflammation/pathology
- Interleukin-6/blood
- Mice, Inbred C57BL
- Monocytes/metabolism
- NFATC Transcription Factors/antagonists & inhibitors
- NFATC Transcription Factors/metabolism
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Pyrazoles/pharmacokinetics
- Pyrazoles/pharmacology
- Signal Transduction/drug effects
- Transcription, Genetic/drug effects
Collapse
Affiliation(s)
| | - Lisa M. Berglund
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Fabiana Blanco
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Eliana Garcia-Vaz
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Maria Wigren
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Pontus Dunér
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | | | - Fong To
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Peter Spegel
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Jan Nilsson
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Eva Bengtsson
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Maria F. Gomez
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
- * E-mail:
| |
Collapse
|
29
|
Li P, Liu Y, Yi B, Wang G, You X, Zhao X, Summer R, Qin Y, Sun J. MicroRNA-638 is highly expressed in human vascular smooth muscle cells and inhibits PDGF-BB-induced cell proliferation and migration through targeting orphan nuclear receptor NOR1. Cardiovasc Res 2013; 99:185-93. [PMID: 23554459 DOI: 10.1093/cvr/cvt082] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIMS Aberrant vascular smooth muscle cell (VSMC) proliferation and migration contribute significantly to the development of vascular pathologies, such as atherosclerosis and restenosis. MicroRNAs have recently emerged as critical modulators in cellular processes and the purpose of this study is to identify novel miRNA regulators implicated in human aortic VSMC proliferation and migration. METHODS AND RESULTS To identify miRNAs that are differentially expressed in human VSMCs, we performed miRNA microarray analysis in human aortic smooth muscle cells (SMCs) at different time points after platelet-derived growth factor (PDGF) stimulation. Here, we identified microRNA-638 (miR-638) as a transcript that was one of the most significantly down-regulated in human VSMCs after PDGF stimulation. Furthermore, we confirmed, by Quantitative RT-PCR, that miR-638 is highly expressed in human VSMCs, and its expression is markedly down-regulated in a dose- and time-dependent manner upon PDGF treatment. Consistent with a critical role in SMC proliferation, we found that miR-638 expression was significantly up-regulated in human VSMCs cultured in differentiation medium, a condition that inhibits SMC proliferation. Furthermore, we identified the orphan nuclear receptor NOR1 as a downstream target gene product of miR-638 and down-regulation of NOR1 is critical for miR-638-mediated inhibitory effects on PDGF-induced cyclin D1 expression, cell proliferation, and migration in human aortic SMCs. CONCLUSION These results indicate that miR-638 is a key molecule in regulating human VSMC proliferation and migration by targeting the NOR1/cyclin D pathway and suggest that specific modulation of miR-638 in human VSMCs may represent an attractive approach for the treatment of proliferative vascular diseases.
Collapse
Affiliation(s)
- Pan Li
- Department of Cardiology, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai 200433, China
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Efficient transduction of vascular smooth muscle cells with a translational AAV2.5 vector: a new perspective for in-stent restenosis gene therapy. Gene Ther 2013; 20:901-12. [PMID: 23535897 PMCID: PMC3706517 DOI: 10.1038/gt.2013.13] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 01/10/2013] [Accepted: 02/05/2013] [Indexed: 12/16/2022]
Abstract
Coronary artery disease represents the leading cause of mortality in the developed world. Percutaneous coronary intervention (PCI) involving stent placement remains disadvantaged by restenosis or thrombosis. Vascular gene-therapy-based methods may be approached, but lack a vascular gene delivery vector. We report a safe and efficient long-term transduction of rat carotid vessels after balloon-injury intervention with a translational optimized AAV2.5 vector. Compared to other known AAV serotypes, AAV2.5 demonstrated the highest transduction efficiency of human coronary artery vascular smooth muscle cells (VSMC) in vitro. Local delivery of AAV2.5-driven transgenes in injured carotid arteries resulted in transduction as soon as day 2 after surgery and persisted for at least 30 days. In contrast to adenovirus 5 vector, inflammation was not detected in AAV2.5-transduced vessels. The functional effects of AAV2.5-mediated gene transfer on neointimal thickening were assessed using the sarco/endoplasmic reticulum Ca2+ ATPase (SERCA2a) human gene, known to inhibit VSMC proliferation. At 30 days, human SERCA2a mRNA was detected in transduced arteries. Morphometric analysis revealed a significant decrease of neointimal hyperplasia in AAV2.5-SERCA2a transduced arteries: 28.36±11.30 (n=8) vs 77.96±24.60 (n=10) μm2, in AAV2.5-GFP-infected, p<0.05. In conclusion, AAV2.5 vector can be considered as a promising safe and effective vector for vascular gene therapy.
Collapse
|
31
|
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]
|
32
|
Abdul-Sater Z, Yehya A, Beresian J, Salem E, Kamar A, Baydoun S, Shibbani K, Soubra A, Bitar F, Nemer G. Two heterozygous mutations in NFATC1 in a patient with Tricuspid Atresia. PLoS One 2012; 7:e49532. [PMID: 23226213 PMCID: PMC3511479 DOI: 10.1371/journal.pone.0049532] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 10/10/2012] [Indexed: 11/19/2022] Open
Abstract
Tricuspid Atresia (TA) is a rare form of congenital heart disease (CHD) with usually poor prognosis in humans. It presents as a complete absence of the right atrio-ventricular connection secured normally by the tricuspid valve. Defects in the tricuspid valve are so far not associated with any genetic locus, although mutations in numerous genes were linked to multiple forms of congenital heart disease. In the last decade, Knock-out mice have offered models for cardiologists and geneticists to study the causes of congenital disease. One such model was the Nfatc1(-/-) mice embryos which die at mid-gestation stage due to a complete absence of the valves. NFATC1 belongs to the Rel family of transcription factors members of which were shown to be implicated in gene activation, cell differentiation, and organogenesis. We have previously shown that a tandem repeat in the intronic region of NFATC1 is associated with ventricular septal defects. In this report, we unravel for the first time a potential link between a mutation in NFATC1 and TA. Two heterozygous missense mutations were found in the NFATC1 gene in one indexed-case out of 19 patients with TA. The two amino-acids changes were not found neither in other patients with CHDs, nor in the control healthy population. Moreover, we showed that these mutations alter dramatically the normal function of the protein at the cellular localization, DNA binding and transcriptional levels suggesting they are disease-causing.
Collapse
Affiliation(s)
- Zahi Abdul-Sater
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Amin Yehya
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Jean Beresian
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Elie Salem
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Amina Kamar
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Serine Baydoun
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Kamel Shibbani
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Ayman Soubra
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Fadi Bitar
- Department of Pediatrics and Adolescent Medicine, American University of Beirut, Beirut, Lebanon
| | - Georges Nemer
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
33
|
Singh NK, Kundumani-Sridharan V, Kumar S, Verma SK, Kotla S, Mukai H, Heckle MR, Rao GN. Protein kinase N1 is a novel substrate of NFATc1-mediated cyclin D1-CDK6 activity and modulates vascular smooth muscle cell division and migration leading to inward blood vessel wall remodeling. J Biol Chem 2012; 287:36291-304. [PMID: 22893700 DOI: 10.1074/jbc.m112.361220] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Toward understanding the mechanisms of vascular wall remodeling, here we have studied the role of NFATc1 in MCP-1-induced human aortic smooth muscle cell (HASMC) growth and migration and injury-induced rat aortic wall remodeling. We have identified PKN1 as a novel downstream target of NFATc1-cyclin D1/CDK6 activity in mediating vascular wall remodeling following injury. MCP-1, a potent chemoattractant protein, besides enhancing HASMC motility, also induced its growth, and these effects require NFATc1-dependent cyclin D1 expression and CDK4/6 activity. In addition, MCP-1 induced PKN1 activation in a sustained and NFATc1-cyclin D1/CDK6-dependent manner. Furthermore, PKN1 activation is required for MCP-1-induced HASMC growth and migration. Balloon injury induced PKN1 activation in NFAT-dependent manner and pharmacological or dominant negative mutant-mediated blockade of PKN1 function or siRNA-mediated down-regulation of its levels substantially suppressed balloon injury-induced smooth muscle cell migration and proliferation resulting in reduced neointima formation. These novel findings suggest that PKN1 plays a critical role in vascular wall remodeling, and therefore, it could be a promising new target for the next generation of drugs for vascular diseases, particularly restenosis following angioplasty, stent implantation, or vein grafting.
Collapse
Affiliation(s)
- Nikhlesh K Singh
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Wang YS, Wang HYJ, Liao YC, Tsai PC, Chen KC, Cheng HY, Lin RT, Juo SHH. MicroRNA-195 regulates vascular smooth muscle cell phenotype and prevents neointimal formation. Cardiovasc Res 2012; 95:517-26. [PMID: 22802111 DOI: 10.1093/cvr/cvs223] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
AIMS Proliferation and migration of vascular smooth muscle cells (VSMCs) can cause atherosclerosis and neointimal formation. MicroRNAs have been shown to regulate cell proliferation and phenotype transformation. We discovered abundant expression of microRNA-195 in VSMCs and conducted a series of studies to identify its function in the cardiovascular system. METHODS AND RESULTS MicroRNA-195 expression was initially found to be altered when VSMCs were treated with oxidized low-density lipoprotein (oxLDL) in a non-replicated microRNA array experiment. Using cellular studies, we found that microRNA-195 reduced VSMC proliferation, migration, and synthesis of IL-1β, IL-6, and IL-8. Using bioinformatics prediction and experimental studies, we showed that microRNA-195 could repress the expression of Cdc42, CCND1, and FGF1 genes. Using a rat model, we found that the microRNA-195 gene, introduced by adenovirus, substantially reduced neointimal formation in a balloon-injured carotid artery. In situ hybridization confirmed the presence of microRNA-195 in the treated arteries but not in control arteries. Immunohistochemistry experiments showed abundant Cdc42 in the neointima of treated arteries. CONCLUSIONS We showed that microRNA-195 plays a role in the cardiovascular system by inhibiting VSMC proliferation, migration, and proinflammatory biomarkers. MicroRNA-195 may have the potential to reduce neointimal formation in patients receiving stenting or angioplasty.
Collapse
Affiliation(s)
- Yung-Song Wang
- Department of Genome Medicine, Kaohsiung Medical University, No. 100, TzYou First Road, Kaohsiung 80708, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Kundumani-Sridharan V, Van Quyen D, Subramani J, Singh NK, Chin YE, Rao GN. Novel interactions between NFATc1 (Nuclear Factor of Activated T cells c1) and STAT-3 (Signal Transducer and Activator of Transcription-3) mediate G protein-coupled receptor agonist, thrombin-induced biphasic expression of cyclin D1, with first phase influencing cell migration and second phase directing cell proliferation. J Biol Chem 2012; 287:22463-82. [PMID: 22566696 DOI: 10.1074/jbc.m112.362996] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Thrombin, a G protein-coupled receptor agonist, induced a biphasic expression of cyclin D1 in primary vascular smooth muscle cells. Although both phases of cyclin D1 expression require binding of the newly identified cooperative complex, NFATc1·STAT-3, to its promoter, the second phase, which is more robust, depends on NFATc1-mediated recruitment of p300 onto the complex and the subsequent acetylation of STAT-3. In addition, STAT-3 is tyrosine-phosphorylated in a biphasic manner, and the late phase requires NFATc1-mediated p300-dependent acetylation. Furthermore, interference with acetylation of STAT-3 by overexpression of acetylation null STAT-3 mutant led to the loss of the late phase of cyclin D1 expression. EMSA analysis and reporter gene assays revealed that NFATc1·STAT-3 complex binding to the cyclin D1 promoter led to an enhanceosome formation and facilitated cyclin D1 expression. In the early phase of its expression, cyclin D1 is localized mostly in the cytoplasm and influenced cell migration. However, during the late and robust phase of its expression, cyclin D1 is translocated to the nucleus and directed cell proliferation. Together, these results demonstrate for the first time that the dual function of cyclin D1 in cell migration and proliferation is temperospatially separated by its biphasic expression, which is mediated by cooperative interactions between NFATc1 and STAT-3.
Collapse
|
36
|
Wong CK, So WY, Law SK, Leung FP, Yau KL, Yao X, Huang Y, Li X, Tsang SY. Estrogen controls embryonic stem cell proliferation via store-operated calcium entry and the nuclear factor of activated T-cells (NFAT). J Cell Physiol 2012; 227:2519-30. [PMID: 21898397 DOI: 10.1002/jcp.22990] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Embryonic stem cells (ESCs) can self-renew indefinitely and differentiate into all cell lineages. Calcium is a universal second messenger which regulates a number of cellular pathways. Previous studies showed that store-operated calcium channels (SOCCs) but not voltage-operated calcium channels are present in mouse ESCs (mESCs). In this study, store-operated calcium entry (SOCE) was found to exist in mESCs using confocal microscopy. SOCC blockers lanthanum, 2-aminoethoxydiphenyl borate (2-APB) and SKF-96365 reduced mESC proliferation in a concentration-dependent manner, suggesting that SOCE is important for ESC proliferation. Pluripotent markers, Sox-2, Klf-4, and Nanog, were down-regulated by 2-APB, suggesting that self-renewal property of mESCs relies on SOCE. 17β-estradiol (E2) enhanced mESC proliferation. This enhanced proliferation was associated with an increment of SOCE. Both stimulated proliferation and increased SOCE could be reversed by SOCC blockers suggesting that E2 mediates its stimulatory effect on proliferation via enhancing SOCE. Also, cyclosporin A and INCA-6, inhibitors of calcineurin [phosphatase that de-phosphorylates and activates nuclear factor of activated T-cells (NFAT)], reversed the proliferative effect of E2, indicating that NFAT is involved in E2-stimulated proliferation. Interestingly, E2 caused the nuclear translocation of NFATc4, and this could be reversed by 2-APB. These results suggested that NFATc4 is the downstream target of E2-induced SOCE. The present investigation provides the first line of evidence that SOCE and NFAT are crucial for ESCs to maintain their unique characteristics. In addition, the present investigation also provides novel information on the mechanisms of how E2, an important female sex hormone, affects ESC proliferation.
Collapse
Affiliation(s)
- Chun-Kit Wong
- School of Life Sciences, Chinese University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Guan H, Zhu L, Fu M, Yang D, Tian S, Guo Y, Cui C, Wang L, Jiang H. 3,3'Diindolylmethane suppresses vascular smooth muscle cell phenotypic modulation and inhibits neointima formation after carotid injury. PLoS One 2012; 7:e34957. [PMID: 22506059 PMCID: PMC3323601 DOI: 10.1371/journal.pone.0034957] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 03/08/2012] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND 3,3'Diindolylmethane (DIM), a natural phytochemical, has shown inhibitory effects on the growth and migration of a variety of cancer cells; however, whether DIM has similar effects on vascular smooth muscle cells (VSMCs) remains unknown. The purpose of this study was to assess the effects of DIM on the proliferation and migration of cultured VSMCs and neointima formation in a carotid injury model, as well as the related cell signaling mechanisms. METHODOLOGY/PRINCIPAL FINDINGS DIM dose-dependently inhibited the platelet-derived growth factor (PDGF)-BB-induced proliferation of VSMCs without cell cytotoxicity. This inhibition was caused by a G0/G1 phase cell cycle arrest demonstrated by fluorescence-activated cell-sorting analysis. We also showed that DIM-induced growth inhibition was associated with the inhibition of the expression of cyclin D1 and cyclin-dependent kinase (CDK) 4/6 as well as an increase in p27(Kip1) levels in PDGF-stimulated VSMCs. Moreover, DIM was also found to modulate migration of VSMCs and smooth muscle-specific contractile marker expression. Mechanistically, DIM negatively modulated PDGF-BB-induced phosphorylation of PDGF-recptorβ (PDGF-Rβ) and the activities of downstream signaling molecules including Akt/glycogen synthase kinase(GSK)3β, extracellular signal-regulated kinase1/2 (ERK1/2), and signal transducers and activators of transcription 3 (STAT3). Our in vivo studies using a mouse carotid arterial injury model revealed that treatment with 150 mg/kg DIM resulted in significant reduction of the neointima/media ratio and proliferating cell nuclear antigen (PCNA)-positive cells, without affecting apoptosis of vascular cells and reendothelialization. Infiltration of inflammatory cells was also inhibited by DIM administration. CONCLUSION These results demonstrate that DIM can suppress the phenotypic modulation of VSMCs and neointima hyperplasia after vascular injury. These beneficial effects on VSMCs were at least partly mediated by the inhibition of PDGF-Rβ and the activities of downstream signaling pathways. The results suggest that DIM has the potential to be a candidate for the prevention of restenosis.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Becaplermin
- Carotid Artery Diseases/drug therapy
- Carotid Artery Diseases/metabolism
- Carotid Artery Diseases/pathology
- Cell Cycle Checkpoints/drug effects
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Cyclin D1/metabolism
- Cyclin-Dependent Kinases/metabolism
- G1 Phase/drug effects
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/metabolism
- Human Umbilical Vein Endothelial Cells/pathology
- Humans
- Indoles/pharmacology
- Inflammation/drug therapy
- Inflammation/metabolism
- Male
- Mice
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima/drug therapy
- Neointima/metabolism
- Neointima/pathology
- Phosphorylation/drug effects
- Proliferating Cell Nuclear Antigen/metabolism
- Proto-Oncogene Proteins c-sis/metabolism
- Rats, Sprague-Dawley
- Resting Phase, Cell Cycle/drug effects
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Hongjing Guan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute of Wuhan University, Wuhan, People's Republic of China
| | - Lihua Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute of Wuhan University, Wuhan, People's Republic of China
| | - Mingyue Fu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute of Wuhan University, Wuhan, People's Republic of China
| | - Da Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute of Wuhan University, Wuhan, People's Republic of China
| | - Song Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute of Wuhan University, Wuhan, People's Republic of China
| | - Yuanyuan Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute of Wuhan University, Wuhan, People's Republic of China
| | - Changping Cui
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute of Wuhan University, Wuhan, People's Republic of China
| | - Lang Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute of Wuhan University, Wuhan, People's Republic of China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute of Wuhan University, Wuhan, People's Republic of China
- * E-mail:
| |
Collapse
|
38
|
Mill C, George SJ. Wnt signalling in smooth muscle cells and its role in cardiovascular disorders. Cardiovasc Res 2012; 95:233-40. [PMID: 22492675 DOI: 10.1093/cvr/cvs141] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Vascular smooth muscle cells (SMCs) are the major cell type within blood vessels. SMCs exhibit low rates of proliferation, migration, and apoptosis in normal blood vessels. However, increased SMC proliferation, migration, and apoptosis rates radically alter the composition and structure of the blood vessel wall and contribute to cardiovascular diseases, such as atherosclerosis, and restenosis that occur after coronary artery vein grafting and stent implantation. Consequently, therapies that modulate SMC proliferation, migration, and apoptosis may be useful for treating cardiovascular diseases. The family of Wnt proteins, which were first identified in the wingless drosophila, has a well-established role in embryogenesis and development. It is now emerging that Wnt proteins also regulate SMC proliferation, migration, and survival. In this review article, we discuss recently emerging research that has revealed that Wnt proteins are important regulators of SMC behaviour via activation of β-catenin-dependent and β-catenin-independent Wnt signalling pathways.
Collapse
Affiliation(s)
- Carina Mill
- Bristol Heart Institute, School of Clinical Sciences, Research Floor Level 7, Bristol Royal Infirmary, Upper Maudlin St, Bristol BS2 8HW, UK
| | | |
Collapse
|
39
|
Daniel JM, Dutzmann J, Bielenberg W, Widmer-Teske R, Gündüz D, Hamm CW, Sedding DG. Inhibition of STAT3 signaling prevents vascular smooth muscle cell proliferation and neointima formation. Basic Res Cardiol 2012; 107:261. [PMID: 22418922 PMCID: PMC3350628 DOI: 10.1007/s00395-012-0261-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 02/22/2012] [Accepted: 03/07/2012] [Indexed: 12/11/2022]
Abstract
Dedifferentiation, migration, and proliferation of resident vascular smooth muscle cells (SMCs) are key components of neointima formation after vascular injury. Activation of signal transducer and activator of transcription-3 (STAT3) is suggested to be critically involved in this process, but the complex regulation of STAT3-dependent genes and the functional significance of inhibiting this pathway during the development of vascular proliferative diseases remain elusive. In this study, we demonstrate that STAT3 was activated in neointimal lesions following wire-induced injury in mice. Phosphorylation of STAT3 induced trans-activation of cyclin D1 and survivin in SMCs in vitro and in neointimal cells in vivo, thus promoting proliferation and migration of SMCs as well as reducing apoptotic cell death. WP1066, a highly potent inhibitor of STAT3 signaling, abrogated phosphorylation of STAT3 and dose-dependently inhibited the functional effects of activated STAT3 in stimulated SMCs. The local application of WP1066 via a thermosensitive pluronic F-127 gel around the dilated arteries significantly inhibited proliferation of neointimal cells and decreased the neointimal lesion size at 3 weeks after injury. Even though WP1066 application attenuated the injury-induced up-regulation of the chemokine RANTES at 6 h after injury, there was no significant effect on the accumulation of circulating cells at 1 week after injury. In conclusion, these data identify STAT3 as a key molecule for the proliferative response of SMC and neointima formation. Moreover, inhibition of STAT3 by the potent and specific compound WP1066 might represent a novel and attractive approach for the local treatment of vascular proliferative diseases.
Collapse
Affiliation(s)
- Jan-Marcus Daniel
- Department of Cardiology, Justus-Liebig-University, Giessen, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Guan H, Gao L, Zhu L, Yan L, Fu M, Chen C, Dong X, Wang L, Huang K, Jiang H. Apigenin attenuates neointima formation via suppression of vascular smooth muscle cell phenotypic transformation. J Cell Biochem 2012; 113:1198-207. [DOI: 10.1002/jcb.23452] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
41
|
Lee MY, Garvey SM, Ripley ML, Wamhoff BR. Genome-wide microarray analyses identify the protein C receptor as a novel calcineurin/nuclear factor of activated T cells-dependent gene in vascular smooth muscle cell phenotypic modulation. Arterioscler Thromb Vasc Biol 2012; 31:2665-75. [PMID: 21903947 DOI: 10.1161/atvbaha.111.235960] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Calcineurin (Cn) and the nuclear factor of activated T cells (NFAT) family of transcription factors are critical in vascular smooth muscle cell (SMC) development and pathology. Here, we used a genomics approach to identify and validate NFAT gene targets activated during platelet-derived growth factor-BB (PDGF-BB)-induced SMC phenotypic modulation. METHODS AND RESULTS Genome-wide expression arrays were used to identify genes both (1) differentially activated in response to PDGF-BB and (2) whose differential expression was reduced by both the Cn inhibitor cyclosporin A and the NFAT inhibitor A-285222. The 20 most pharmacologically sensitive genes were validated by quantitative reverse transcription-polymerase chain reaction analysis of PDGF-BB-stimulated SMCs in the presence of Cn/NFAT inhibitors, including the VIVIT peptide. In all experiments, protein C receptor (PROCR) gene activation was reduced. We showed that PROCR expression was virtually absent in untreated, quiescent SMCs. PDGF-BB stimulation, however, induced significant PROCR promoter activation and downstream protein expression in a Cn/NFAT-dependent manner. Mutation of a species-conserved, NFAT binding motif significantly attenuated PDGF-BB-induced PROCR promoter activity, thereby distinguishing NFAT as the first PROCR transcriptional activator to date. Moreover, SMC PROCR expression was upregulated in the neointima as early as 7 days following acute vascular injury in rat carotid arteries. CONCLUSION We hereby report PROCR as a novel, NFAT-dependent gene that may be implicated in vascular restenosis and consequent inward remodeling.
Collapse
Affiliation(s)
- Monica Y Lee
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | |
Collapse
|
42
|
Calcium signaling in vascular smooth muscle cells: from physiology to pathology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:795-810. [PMID: 22453970 DOI: 10.1007/978-94-007-2888-2_35] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cyclic variations in calcium (Ca(2+)) concentrations, through a process called excitation-contraction coupling, allow regulation of vascular smooth muscle cells contractility and thus modulation of vascular tone and blood pressure. As a second messenger, Ca(2+) also activates signaling cascades leading to transcription factors activation in a process called excitation-transcription coupling. Furthermore, recent evidences indicate an interaction between post-transcriptional regulation by microRNAs (miRNAs) and Ca(2+) signaling. All these actors, which are frequently altered in vascular diseases, will be reviewed here.
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW We aim to highlight the emerging evidence for the role of the Wnt signalling pathways in vascular disease and indicate how our current understanding is supported by observations of Wnt signalling in vascular development. RECENT FINDINGS There is mounting direct and indirect evidence for an involvement of the Wnt pathways in multiple processes involved in atherogenesis. Although a systematic analysis of Wnt pathway in atherosclerosis has not been performed, it is apparent that altered expression of a handful of Wnt pathway proteins occurs in or regulates atherogenesis. Wnt pathways regulate endothelial dysfunction and vascular smooth muscle cell (VSMC) proliferation and migration and thereby intimal thickening. Furthermore, the Wnt pathways have the capacity to regulate inflammation and foam cell formation, pathological angiogenesis and calcification, which are crucial processes in plaque formation and stability. SUMMARY A wealth of evidence has been presented for the involvement of the Wnt pathways in vascular development. Although less evidence exists for the regulation of vascular disease by the Wnt pathways, sufficient evidence exists to propose these pathways act as an important regulator of vascular disease. A greater understanding of Wnt pathways may reveal new therapeutic targets for vascular disease.
Collapse
Affiliation(s)
- Aikaterini Tsaousi
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Research Floor Level Seven, Bristol Royal Infirmary, Bristol, UK
| | | | | |
Collapse
|
44
|
Goettsch C, Rauner M, Hamann C, Sinningen K, Hempel U, Bornstein SR, Hofbauer LC. Nuclear factor of activated T cells mediates oxidised LDL-induced calcification of vascular smooth muscle cells. Diabetologia 2011; 54:2690-701. [PMID: 21701818 DOI: 10.1007/s00125-011-2219-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 05/23/2011] [Indexed: 12/21/2022]
Abstract
AIMS/HYPOTHESIS Vascular calcification is a prominent feature of both atherosclerosis and diabetes, and is clinically associated with osteoporosis. The expression of bone-regulatory factors and the impact of oxidative stress in aortic calcification are well-documented. Recently, nuclear factor of activated T cells (NFAT) cytoplasmic, calcineurin-dependent 1 (NFATc1) was identified in calcified aortic valves and has been implicated in vascular calcification. Therefore, we assessed the mechanisms of osteogenic transdifferentiation of vascular smooth muscle cells induced by oxidised LDL (oxLDL) and evaluated the role of NFAT in this process. METHODS Human coronary artery smooth muscle cells (HCASMCs) were cultured for 21 days in medium supplemented with oxLDL. NFAT was inhibited using the NFAT inhibitor VIVIT, or by knockdown with small interfering RNA (siRNA). Osteogenic transdifferentiation was assessed by gene expression, matrix mineralisation and alkaline phosphatase activity. RESULTS Exposure to oxLDL caused the transformation of HCASMCs towards an osteoblast-like phenotype based on increased mineral matrix formation and RUNX2 expression. NFATc1 blockade completely prevented oxLDL-induced osteogenic transformation of HCASMCs as well as oxLDL-induced stimulation of osteoblast differentiation. In contrast, matrix mineralisation induced by osteogenic medium was independent of the NFAT pathway. Of note, oxLDL-conditioned medium from HCASMCs transferred to bone cells promoted osteoblast mineralisation. Consistent with these in vitro findings, diabetic rats with a twofold increase in oxidised lipid levels displayed higher aortic calcium concentrations and increased expression of osteogenic markers and production of NFATc1. CONCLUSIONS/INTERPRETATION Our results identify the NFAT signalling pathway as a novel regulator of oxLDL-induced transdifferentiation of vascular smooth muscle cells towards an osteoblast-like phenotype.
Collapse
Affiliation(s)
- C Goettsch
- Division of Endocrinology, Diabetes and Metabolic Bone Diseases, Department of Medicine III, Technical University Medical Center, Fetscherstraße 74, 01307 Dresden, Germany
| | | | | | | | | | | | | |
Collapse
|
45
|
Peyton KJ, Yu Y, Yates B, Shebib AR, Liu XM, Wang H, Durante W. Compound C inhibits vascular smooth muscle cell proliferation and migration in an AMP-activated protein kinase-independent fashion. J Pharmacol Exp Ther 2011; 338:476-84. [PMID: 21566210 DOI: 10.1124/jpet.111.181784] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
6-[4-(2-Piperidin-1-yl-ethoxy)-phenyl]-3-pyridin-4-yl-pyrazolo[1,5-a] pyrimidine (compound C) is a cell-permeable pyrrazolopyrimidine derivative that acts as a potent inhibitor of AMP-activated protein kinase (AMPK). Although compound C is often used to determine the role of AMPK in various physiological processes, it also evokes AMPK-independent actions. In the present study, we investigated whether compound C influences vascular smooth muscle cell (SMC) function through the AMPK pathway. Treatment of rat aortic SMCs with compound C (0.02-10 μM) inhibited vascular SMC proliferation and migration in a concentration-dependent fashion. These actions of compound C were not mimicked or affected by silencing AMPKα expression or infecting SMCs with an adenovirus expressing a dominant-negative mutant of AMPK. In contrast, the pharmacological activator of AMPK 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside inhibited the proliferation and migration of SMCs in a manner that was strictly dependent on AMPK activity. Flow cytometry experiments revealed that compound C arrested SMCs in the G(0)/G(1) phase of the cell cycle, and this was associated with a decrease in cyclin D1 and cyclin A protein expression and retinoblastoma protein phosphorylation and an increase in p21 protein expression. Finally, local perivascular delivery of compound C immediately after balloon injury of rat carotid arteries markedly attenuated neointima formation. These studies identify compound C as a novel AMPK-independent regulator of vascular SMC function that exerts inhibitory effects on SMC proliferation and migration and neointima formation after arterial injury. Compound C represents a potentially new therapeutic agent in treating and preventing occlusive vascular disease.
Collapse
Affiliation(s)
- Kelly J Peyton
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, One Hospital Drive, Columbia, MO 65212, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Guo X, Zhou C, Sun N. The neuropeptide catestatin promotes vascular smooth muscle cell proliferation through the Ca2+-calcineurin-NFAT signaling pathway. Biochem Biophys Res Commun 2011; 407:807-12. [PMID: 21443864 DOI: 10.1016/j.bbrc.2011.03.104] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 03/22/2011] [Indexed: 12/12/2022]
Abstract
The Chromogranin A-derived neuropeptide catestatin is an endogenous nicotinic cholinergic antagonist that acts as a pleiotropic hormone. Since catestatin shares several functions with other members derived from the chromogranin/secretogranin protein family and other neuropeptides which exert proliferative effects on vascular smooth muscle cells (VSMCs), we therefore hypothesized that catestatin would regulate VSMC proliferation. The present study demonstrates that catestatin caused a dose-dependent induction of proliferation in rat aortic smooth muscle cells and furthermore evoked a sustained increase in intracellular calcium. This subsequently leaded to enhanced activation of the Ca(2+)/calmodulin-dependent phosphatase, calcineurin and resulted in an activation of the Ca(2+)-dependent transcription factor, nuclear factor of activated T cells (NFAT), initiating transcription of proliferative genes. In addition, cyclosporin A (CsA), a potent inhibitor of calcineurin, abrogated catestatin-mediated effect on VSMCs, indicating that the calcineurin-NFAT signaling is strongly required for catestatin-induced growth of VSMCs. The present study establishes catestatin as a novel proliferative cytokine on vascular smooth muscle cells and this effect is mediated by the Ca(2+)-calcineurin-NFAT signaling pathway.
Collapse
Affiliation(s)
- Xiaoxia Guo
- Department of Cardiology, People's Hospital, Peking University, No. 11 South Avenue, Xi Zhi Men Xicheng District, Beijing 100044, China
| | | | | |
Collapse
|
47
|
Findeisen HM, Gizard F, Zhao Y, Qing H, Heywood EB, Jones KL, Cohn D, Bruemmer D. Epigenetic regulation of vascular smooth muscle cell proliferation and neointima formation by histone deacetylase inhibition. Arterioscler Thromb Vasc Biol 2011; 31:851-60. [PMID: 21233448 DOI: 10.1161/atvbaha.110.221952] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Proliferation of smooth muscle cells (SMC) in response to vascular injury is central to neointimal vascular remodeling. There is accumulating evidence that histone acetylation constitutes a major epigenetic modification for the transcriptional control of proliferative gene expression; however, the physiological role of histone acetylation for proliferative vascular disease remains elusive. METHODS AND RESULTS In the present study, we investigated the role of histone deacetylase (HDAC) inhibition in SMC proliferation and neointimal remodeling. We demonstrate that mitogens induce transcription of HDAC 1, 2, and 3 in SMC. Short interfering RNA-mediated knockdown of either HDAC 1, 2, or 3 and pharmacological inhibition of HDAC prevented mitogen-induced SMC proliferation. The mechanisms underlying this reduction of SMC proliferation by HDAC inhibition involve a growth arrest in the G(1) phase of the cell cycle that is due to an inhibition of retinoblastoma protein phosphorylation. HDAC inhibition resulted in a transcriptional and posttranscriptional regulation of the cyclin-dependent kinase inhibitors p21(Cip1) and p27(Kip). Furthermore, HDAC inhibition repressed mitogen-induced cyclin D1 mRNA expression and cyclin D1 promoter activity. As a result of this differential cell cycle-regulatory gene expression by HDAC inhibition, the retinoblastoma protein retains a transcriptional repression of its downstream target genes required for S phase entry. Finally, we provide evidence that these observations are applicable in vivo by demonstrating that HDAC inhibition decreased neointima formation and expression of cyclin D1 in a murine model of vascular injury. CONCLUSIONS These findings identify HDAC as a critical component of a transcriptional cascade regulating SMC proliferation and suggest that HDAC might play a pivotal role in the development of proliferative vascular diseases, including atherosclerosis and in-stent restenosis.
Collapse
Affiliation(s)
- Hannes M Findeisen
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Ding X, He Z, Shi Y, Wang Q, Wang Y. Targeting TRPC6 channels in oesophageal carcinoma growth. Expert Opin Ther Targets 2010; 14:513-27. [PMID: 20235901 DOI: 10.1517/14728221003733602] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|