1
|
Bankell E, Liu L, van der Horst J, Rippe C, Jepps TA, Nilsson BO, Swärd K. Suppression of smooth muscle cell inflammation by myocardin-related transcription factors involves inactivation of TANK-binding kinase 1. Sci Rep 2024; 14:13321. [PMID: 38858497 PMCID: PMC11164896 DOI: 10.1038/s41598-024-63901-3] [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: 01/11/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024] Open
Abstract
Myocardin-related transcription factors (MRTFs: myocardin/MYOCD, MRTF-A/MRTFA, and MRTF-B/MRTFB) suppress production of pro-inflammatory cytokines and chemokines in human smooth muscle cells (SMCs) through sequestration of RelA in the NF-κB complex, but additional mechanisms are likely involved. The cGAS-STING pathway is activated by double-stranded DNA in the cytosolic compartment and acts through TANK-binding kinase 1 (TBK1) to spark inflammation. The present study tested if MRTFs suppress inflammation also by targeting cGAS-STING signaling. Interrogation of a transcriptomic dataset where myocardin was overexpressed using a panel of 56 cGAS-STING cytokines showed the panel to be repressed. Moreover, MYOCD, MRTFA, and SRF associated negatively with the panel in human arteries. RT-qPCR in human bronchial SMCs showed that all MRTFs reduced pro-inflammatory cytokines on the panel. MRTFs diminished phosphorylation of TBK1, while STING phosphorylation was marginally affected. The TBK1 inhibitor amlexanox, but not the STING inhibitor H-151, reduced the anti-inflammatory effect of MRTF-A. Co-immunoprecipitation and proximity ligation assays supported binding between MRTF-A and TBK1 in SMCs. MRTFs thus appear to suppress cellular inflammation in part by acting on the kinase TBK1. This may defend SMCs against pro-inflammatory insults in disease.
Collapse
Affiliation(s)
- Elisabeth Bankell
- Cellular Biomechanics/Vascular Physiology, Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden
| | - Li Liu
- Cellular Biomechanics/Vascular Physiology, Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden
- Department of Urology, Qingyuan Hospital Affiliated to Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Jennifer van der Horst
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark
| | - Catarina Rippe
- Cellular Biomechanics/Vascular Physiology, Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden
| | - Thomas A Jepps
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark
| | - Bengt-Olof Nilsson
- Cellular Biomechanics/Vascular Physiology, Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden
| | - Karl Swärd
- Cellular Biomechanics/Vascular Physiology, Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden.
| |
Collapse
|
2
|
Zhang ML, Li HB, Jin Y. Application and perspective of CRISPR/Cas9 genome editing technology in human diseases modeling and gene therapy. Front Genet 2024; 15:1364742. [PMID: 38666293 PMCID: PMC11043577 DOI: 10.3389/fgene.2024.1364742] [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: 01/03/2024] [Accepted: 03/11/2024] [Indexed: 04/28/2024] Open
Abstract
The Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR) mediated Cas9 nuclease system has been extensively used for genome editing and gene modification in eukaryotic cells. CRISPR/Cas9 technology holds great potential for various applications, including the correction of genetic defects or mutations within the human genome. The application of CRISPR/Cas9 genome editing system in human disease research is anticipated to solve a multitude of intricate molecular biology challenges encountered in life science research. Here, we review the fundamental principles underlying CRISPR/Cas9 technology and its recent application in neurodegenerative diseases, cardiovascular diseases, autoimmune related diseases, and cancer, focusing on the disease modeling and gene therapy potential of CRISPR/Cas9 in these diseases. Finally, we provide an overview of the limitations and future prospects associated with employing CRISPR/Cas9 technology for diseases study and treatment.
Collapse
Affiliation(s)
- Man-Ling Zhang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Inner Mongolia Key Laboratory for Pathogenesis and Diagnosis of Rheumatic and Autoimmune Diseases, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Hong-Bin Li
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Inner Mongolia Key Laboratory for Pathogenesis and Diagnosis of Rheumatic and Autoimmune Diseases, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yong Jin
- Department of Rheumatology and Immunology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- Inner Mongolia Key Laboratory for Pathogenesis and Diagnosis of Rheumatic and Autoimmune Diseases, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
3
|
Mann EA, Mogle MS, Park J, Reddy P. Transcription factor Tcf21 modulates urinary bladder size and differentiation. Dev Growth Differ 2024; 66:106-118. [PMID: 38197329 PMCID: PMC11457511 DOI: 10.1111/dgd.12906] [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: 09/12/2023] [Revised: 11/28/2023] [Accepted: 12/12/2023] [Indexed: 01/11/2024]
Abstract
Urinary bladder organogenesis requires coordinated cell growth, specification, and patterning of both mesenchymal and epithelial compartments. Tcf21, a gene that encodes a helix-loop-helix transcription factor, is specifically expressed in the mesenchyme of the bladder during development. Here we show that Tcf21 is required for normal development of the bladder. We found that the bladders of mice lacking Tcf21 were notably hypoplastic and that the Tcf21 mutant mesenchyme showed increased apoptosis. There was also a marked delay in the formation of visceral smooth muscle, accompanied by a defect in myocardin (Myocd) expression. Interestingly, there was also a marked delay in the formation of the basal cell layer of the urothelium, distinguished by diminished expression of Krt5 and Krt14. Our findings suggest that Tcf21 regulates the survival and differentiation of mesenchyme cell-autonomously and the maturation of the adjacent urothelium non-cell-autonomously during bladder development.
Collapse
Affiliation(s)
- Elizabeth A. Mann
- Division of Pediatric UrologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Melissa S. Mogle
- Division of Pediatric UrologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Joo‐Seop Park
- Division of Nephrology and HypertensionNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- The Feinberg Cardiovascular and Renal Research InstituteChicagoIllinoisUSA
| | - Pramod Reddy
- Division of Pediatric UrologyCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| |
Collapse
|
4
|
Liu L, Arévalo-Martínez M, Rippe C, Johansson ME, Holmberg J, Albinsson S, Swärd K. Itga8-Cre-mediated deletion of YAP and TAZ impairs bladder contractility with minimal inflammation and chondrogenic differentiation. Am J Physiol Cell Physiol 2023; 325:C1485-C1501. [PMID: 37927241 DOI: 10.1152/ajpcell.00270.2023] [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: 06/21/2023] [Revised: 10/26/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023]
Abstract
A role of Yes1-associated transcriptional regulator (YAP) and WW domain-containing transcription regulator 1 (TAZ) in vascular and gastrointestinal contractility due to control of myocardin (Myocd) expression, which in turn activates contractile genes, has been demonstrated. Whether this transcriptional hierarchy applies to the urinary bladder is unclear. We found that YAP/TAZ are expressed in human detrusor myocytes and therefore exploited the Itga8-CreERT2 model for the deletion of YAP/TAZ. Recombination occurred in detrusor, and YAP/TAZ transcripts were reduced by >75%. Bladder weights were increased (by ≈22%), but histology demonstrated minimal changes in the detrusor, while arteries in the mucosa were inflamed. Real-time quantitative reverse transcription PCR (RT-qPCR) using the detrusor demonstrated reductions of Myocd (-79 ± 18%) and serum response factor (Srf) along with contractile genes. In addition, the cholinergic receptor muscarinic 2 (Chrm2) and Chrm3 were suppressed (-80 ± 23% and -80 ± 10%), whereas minute increases of Il1b and Il6 were seen. Unlike YAP/TAZ-deficient arteries, SRY (sex-determining region Y)-box 9 (Sox9) did not increase, and no chondrogenic differentiation was apparent. Reductions of smooth muscle myosin heavy chain 11 (Myh11), myosin light-chain kinase gene (Mylk), and Chrm3 were seen at the protein level. Beyond restraining the smooth muscle cell (SMC) program of gene expression, YAP/TAZ depletion silenced SMC-specific splicing, including exon 2a of Myocd. Reduced contractile differentiation was associated with weaker contraction in response to myosin phosphatase inhibition (-36%) and muscarinic activation (reduced by 53% at 0.3 µM carbachol). Finally, short-term overexpression of constitutively active YAP in human embryonic kidney 293 (HEK293) cells increased myocardin (greater than eightfold) along with archetypal target genes, but contractile genes were unaffected or reduced. YAP and TAZ thus regulate myocardin expression in the detrusor, and this is important for SMC differentiation and splicing as well as for contractility.NEW & NOTEWORTHY This study addresses the hypothesis that YAP and TAZ have an overarching role in the transcriptional hierarchy in the smooth muscle of the urinary bladder by controlling myocardin expression. Using smooth muscle-specific and inducible deletion of YAP and TAZ in adult mice, we find that YAP and TAZ control myocardin expression, contractile differentiation, smooth muscle-specific splicing, and bladder contractility. These effects are largely independent of inflammation and chondrogenic differentiation.
Collapse
Affiliation(s)
- Li Liu
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Department of Urology, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | | | - Catarina Rippe
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Martin E Johansson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Johan Holmberg
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Sebastian Albinsson
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Karl Swärd
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
5
|
Arévalo Martínez M, Ritsvall O, Bastrup JA, Celik S, Jakobsson G, Daoud F, Winqvist C, Aspberg A, Rippe C, Maegdefessel L, Schiopu A, Jepps TA, Holmberg J, Swärd K, Albinsson S. Vascular smooth muscle-specific YAP/TAZ deletion triggers aneurysm development in mouse aorta. JCI Insight 2023; 8:e170845. [PMID: 37561588 PMCID: PMC10544211 DOI: 10.1172/jci.insight.170845] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/25/2023] [Indexed: 08/12/2023] Open
Abstract
Inadequate adaption to mechanical forces, including blood pressure, contributes to development of arterial aneurysms. Recent studies have pointed to a mechanoprotective role of YAP and TAZ in vascular smooth muscle cells (SMCs). Here, we identified reduced expression of YAP1 in human aortic aneurysms. Vascular SMC-specific knockouts (KOs) of YAP/TAZ were thus generated using the integrin α8-Cre (Itga8-Cre) mouse model (i8-YT-KO). i8-YT-KO mice spontaneously developed aneurysms in the abdominal aorta within 2 weeks of KO induction and in smaller arteries at later times. The vascular specificity of Itga8-Cre circumvented gastrointestinal effects. Aortic aneurysms were characterized by elastin disarray, SMC apoptosis, and accumulation of proteoglycans and immune cell populations. RNA sequencing, proteomics, and myography demonstrated decreased contractile differentiation of SMCs and impaired vascular contractility. This associated with partial loss of myocardin expression, reduced blood pressure, and edema. Mediators in the inflammatory cGAS/STING pathway were increased. A sizeable increase in SOX9, along with several direct target genes, including aggrecan (Acan), contributed to proteoglycan accumulation. This was the earliest detectable change, occurring 3 days after KO induction and before the proinflammatory transition. In conclusion, Itga8-Cre deletion of YAP and TAZ represents a rapid and spontaneous aneurysm model that recapitulates features of human abdominal aortic aneurysms.
Collapse
Affiliation(s)
| | - Olivia Ritsvall
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Joakim Armstrong Bastrup
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Selvi Celik
- Molecular Cardiology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Gabriel Jakobsson
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Fatima Daoud
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Christopher Winqvist
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Anders Aspberg
- Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Catarina Rippe
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Lars Maegdefessel
- Department of Medicine, Karolinska Institute, Stockholm, Sweden, and
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar - Technical University Munich (TUM), Munich, Germany
| | - Alexandru Schiopu
- Department of Translational Medicine, Lund University, Malmö, Sweden
- Department of Internal Medicine, Skåne University Hospital Lund, Lund, Sweden, and
- Nicolae Simionescu Institute of Cellular Biology and Pathology, Bucharest, Romania
| | - Thomas A. Jepps
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Johan Holmberg
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Karl Swärd
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Sebastian Albinsson
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
6
|
Liu H, Dong X, Jia K, Yuan B, Ren Z, Pan X, Wu J, Li J, Zhou J, Wang RX, Qu L, Sun J, Pan LL. Protein arginine methyltransferase 5-mediated arginine methylation stabilizes Kruppel-like factor 4 to accelerate neointimal formation. Cardiovasc Res 2023; 119:2142-2156. [PMID: 37201513 DOI: 10.1093/cvr/cvad080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 01/28/2023] [Accepted: 03/01/2023] [Indexed: 05/20/2023] Open
Abstract
AIMS Accumulating evidence supports the indispensable role of protein arginine methyltransferase 5 (PRMT5) in the pathological progression of several human cancers. As an important enzyme-regulating protein methylation, how PRMT5 participates in vascular remodelling remains unknown. The aim of this study was to investigate the role and underlying mechanism of PRMT5 in neointimal formation and to evaluate its potential as an effective therapeutic target for the condition. METHODS AND RESULTS Aberrant PRMT5 overexpression was positively correlated with clinical carotid arterial stenosis. Vascular smooth muscle cell (SMC)-specific PRMT5 knockout inhibited intimal hyperplasia with an enhanced expression of contractile markers in mice. Conversely, PRMT5 overexpression inhibited SMC contractile markers and promoted intimal hyperplasia. Furthermore, we showed that PRMT5 promoted SMC phenotypic switching by stabilizing Kruppel-like factor 4 (KLF4). Mechanistically, PRMT5-mediated KLF4 methylation inhibited ubiquitin-dependent proteolysis of KLF4, leading to a disruption of myocardin (MYOCD)-serum response factor (SRF) interaction and MYOCD-SRF-mediated the transcription of SMC contractile markers. CONCLUSION Our data demonstrated that PRMT5 critically mediated vascular remodelling by promoting KLF4-mediated SMC phenotypic conversion and consequently the progression of intimal hyperplasia. Therefore, PRMT5 may represent a potential therapeutic target for intimal hyperplasia-associated vascular diseases.
Collapse
Affiliation(s)
- He Liu
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, No. 1800 Lihu Avenue, Wuxi 214122, P. R. China
- State Key Laboratory of Food Science and Resources, Jiangnan University, No. 1800 Lihu Avenue, Wuxi 214122, P. R. China
| | - Xiaoliang Dong
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, No. 1800 Lihu Avenue, Wuxi 214122, P. R. China
| | - Kunpeng Jia
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, No. 1800 Lihu Avenue, Wuxi 214122, P. R. China
- State Key Laboratory of Food Science and Resources, Jiangnan University, No. 1800 Lihu Avenue, Wuxi 214122, P. R. China
| | - Baohui Yuan
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, No. 1800 Lihu Avenue, Wuxi 214122, P. R. China
- State Key Laboratory of Food Science and Resources, Jiangnan University, No. 1800 Lihu Avenue, Wuxi 214122, P. R. China
| | - Zhengnan Ren
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, No. 1800 Lihu Avenue, Wuxi 214122, P. R. China
- State Key Laboratory of Food Science and Resources, Jiangnan University, No. 1800 Lihu Avenue, Wuxi 214122, P. R. China
| | - Xiaohua Pan
- State Key Laboratory of Food Science and Resources, Jiangnan University, No. 1800 Lihu Avenue, Wuxi 214122, P. R. China
| | - Jianjin Wu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital, Navy Military Medical University, 415 Fengyang Road, Shanghai 200003, P. R. China
| | - Jiahong Li
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, No. 1800 Lihu Avenue, Wuxi 214122, P. R. China
- State Key Laboratory of Food Science and Resources, Jiangnan University, No. 1800 Lihu Avenue, Wuxi 214122, P. R. China
| | - Jingwen Zhou
- National Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, No. 1800 Lihu Avenue, Wuxi 214122, P. R. China
| | - Ru-Xing Wang
- Department of Cardiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, 299 Qingyang Road, Wuxi 214023, P. R. China
| | - Lefeng Qu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital, Navy Military Medical University, 415 Fengyang Road, Shanghai 200003, P. R. China
| | - Jia Sun
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, No. 1800 Lihu Avenue, Wuxi 214122, P. R. China
- State Key Laboratory of Food Science and Resources, Jiangnan University, No. 1800 Lihu Avenue, Wuxi 214122, P. R. China
| | - Li-Long Pan
- Wuxi School of Medicine and School of Food Science and Technology, Jiangnan University, No. 1800 Lihu Avenue, Wuxi 214122, P. R. China
| |
Collapse
|
7
|
Song Y, Wang T, Mu C, Gui W, Deng Y, Ma R. LncRNA SENCR overexpression attenuated the proliferation, migration and phenotypic switching of vascular smooth muscle cells in aortic dissection via the miR-206/myocardin axis. Nutr Metab Cardiovasc Dis 2022; 32:1560-1570. [PMID: 35351345 DOI: 10.1016/j.numecd.2022.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND AIMS Smooth muscle and endothelial cell-enriched migration/differentiation-associated lncRNA (SENCR) has been reported to be associated with some cardiovascular diseases; however, its function and exact molecular mechanism in aortic dissection (AD) remain undefined. Thus, we investigated the effects of SENCR on AD and its potential mechanisms. METHODS AND RESULTS SENCR expression in aortic media specimens from AD patients was detected by quantitative real-time PCR (qPCR). The roles of SENCR in vascular smooth muscle cell (VMSC) proliferation and migration as well as in the regulation of contractile phenotype genes were studied using CCK-8, wound healing, Transwell, qPCR and Western blot assays. Dual-luciferase reporter assays were performed to identify the regulatory correlation between SENCR, miR-206 and myocardin. Furthermore, mouse AD models were constructed with ApoE-/- mice, and the effect of upregulated SENCR on phenotypic switching in the AD model was detected using hematoxylin and eosin (H&E) staining and immunohistochemistry (IHC) assays. SENCR overexpression inhibited VSMC proliferation, migration and synthetic phenotype-related gene expression; decreased miR-206 expression; increased myocardin expression; and suppressed rupture of the aortic media in mice. SENCR knockdown had the opposite effects. Our results further suggested that miR-206 upregulation could reverse the inhibitory roles of SENCR upregulation and that myocardin upregulation could restore the function of SENCR upregulation in VSMCs. Dual-luciferase reporter assays confirmed that SENCR regulated miR-206, which directly targeted myocardin in VSMCs. CONCLUSION SENCR overexpression suppressed VMSC proliferation and migration, maintained the contractile phenotype and suppressed aortic dilatation via the miR-206/myocardin axis.
Collapse
Affiliation(s)
- Yi Song
- Department of Extracorporeal Circulation, Fuwai Yunnan Cardiovascular Hospital, Kunming, 650102, China
| | - Tao Wang
- Department of Cardiothoracic Surgery, The First People's Hospital of Kunming, Kunming, 650034, China
| | - Chunjie Mu
- Department of Cardiovascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, 650102, China
| | - Wenting Gui
- Department of Cardiovascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, 650102, China
| | - Yao Deng
- Department of Cardiovascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, 650102, China
| | - Runwei Ma
- Department of Cardiovascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, 650102, China.
| |
Collapse
|
8
|
Rogawski R, Sharon M. Characterizing Endogenous Protein Complexes with Biological Mass Spectrometry. Chem Rev 2022; 122:7386-7414. [PMID: 34406752 PMCID: PMC9052418 DOI: 10.1021/acs.chemrev.1c00217] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Indexed: 01/11/2023]
Abstract
Biological mass spectrometry (MS) encompasses a range of methods for characterizing proteins and other biomolecules. MS is uniquely powerful for the structural analysis of endogenous protein complexes, which are often heterogeneous, poorly abundant, and refractive to characterization by other methods. Here, we focus on how biological MS can contribute to the study of endogenous protein complexes, which we define as complexes expressed in the physiological host and purified intact, as opposed to reconstituted complexes assembled from heterologously expressed components. Biological MS can yield information on complex stoichiometry, heterogeneity, topology, stability, activity, modes of regulation, and even structural dynamics. We begin with a review of methods for isolating endogenous complexes. We then describe the various biological MS approaches, focusing on the type of information that each method yields. We end with future directions and challenges for these MS-based methods.
Collapse
Affiliation(s)
- Rivkah Rogawski
- Department of Biomolecular
Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Michal Sharon
- Department of Biomolecular
Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
9
|
Dong K, Shen J, He X, Hu G, Wang L, Osman I, Bunting KM, Dixon-Melvin R, Zheng Z, Xin H, Xiang M, Vazdarjanova A, Fulton DJR, Zhou J. CARMN Is an Evolutionarily Conserved Smooth Muscle Cell-Specific LncRNA That Maintains Contractile Phenotype by Binding Myocardin. Circulation 2021; 144:1856-1875. [PMID: 34694145 PMCID: PMC8726016 DOI: 10.1161/circulationaha.121.055949] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Vascular homeostasis is maintained by the differentiated phenotype of vascular smooth muscle cells (VSMCs). The landscape of protein coding genes comprising the transcriptome of differentiated VSMCs has been intensively investigated but many gaps remain including the emerging roles of noncoding genes. METHODS We reanalyzed large-scale, publicly available bulk and single-cell RNA sequencing datasets from multiple tissues and cell types to identify VSMC-enriched long noncoding RNAs. The in vivo expression pattern of a novel smooth muscle cell (SMC)-expressed long noncoding RNA, Carmn (cardiac mesoderm enhancer-associated noncoding RNA), was investigated using a novel Carmn green fluorescent protein knock-in reporter mouse model. Bioinformatics and quantitative real-time polymerase chain reaction analysis were used to assess CARMN expression changes during VSMC phenotypic modulation in human and murine vascular disease models. In vitro, functional assays were performed by knocking down CARMN with antisense oligonucleotides and overexpressing Carmn by adenovirus in human coronary artery SMCs. Carotid artery injury was performed in SMC-specific Carmn knockout mice to assess neointima formation and the therapeutic potential of reversing CARMN loss was tested in a rat carotid artery balloon injury model. The molecular mechanisms underlying CARMN function were investigated using RNA pull-down, RNA immunoprecipitation, and luciferase reporter assays. RESULTS We identified CARMN, which was initially annotated as the host gene of the MIR143/145 cluster and recently reported to play a role in cardiac differentiation, as a highly abundant and conserved, SMC-specific long noncoding RNA. Analysis of the Carmn GFP knock-in mouse model confirmed that Carmn is transiently expressed in embryonic cardiomyocytes and thereafter becomes restricted to SMCs. We also found that Carmn is transcribed independently of Mir143/145. CARMN expression is dramatically decreased by vascular disease in humans and murine models and regulates the contractile phenotype of VSMCs in vitro. In vivo, SMC-specific deletion of Carmn significantly exacerbated, whereas overexpression of Carmn markedly attenuated, injury-induced neointima formation in mouse and rat, respectively. Mechanistically, we found that Carmn physically binds to the key transcriptional cofactor myocardin, facilitating its activity and thereby maintaining the contractile phenotype of VSMCs. CONCLUSIONS CARMN is an evolutionarily conserved SMC-specific long noncoding RNA with a previously unappreciated role in maintaining the contractile phenotype of VSMCs and is the first noncoding RNA discovered to interact with myocardin.
Collapse
Affiliation(s)
- Kunzhe Dong
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
| | - Jian Shen
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Xiangqin He
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
| | - Guoqing Hu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
| | - Liang Wang
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Islam Osman
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
| | - Kristopher M. Bunting
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
| | - Rachael Dixon-Melvin
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
| | - Zeqi Zheng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Hongbo Xin
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330031, China
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, 330031, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Almira Vazdarjanova
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
| | - David J. R. Fulton
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
| | - Jiliang Zhou
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, 30912, USA
| |
Collapse
|
10
|
Gurumurthy CB, Saunders TL, Ohtsuka M. Designing and generating a mouse model: frequently asked questions. J Biomed Res 2021; 35:76-90. [PMID: 33797414 PMCID: PMC8038528 DOI: 10.7555/jbr.35.20200197] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Genetically engineered mouse (GEM) models are commonly used in biomedical research. Generating GEMs involve complex set of experimental procedures requiring sophisticated equipment and highly skilled technical staff. Because of these reasons, most research institutes set up centralized core facilities where custom GEMs are created for research groups. Researchers, on the other hand, when they begin thinking about generating GEMs for their research, several questions arise in their minds. For example, what type of model(s) would be best useful for my research, how do I design them, what are the latest technologies and tools available for developing my model(s), and finally how to breed GEMs in my research. As there are several considerations and options in mouse designs, and as it is an expensive and time-consuming endeavor, careful planning upfront can ensure the highest chance of success. In this article, we provide brief answers to several frequently asked questions that arise when researchers begin thinking about generating mouse model(s) for their work.
Collapse
Affiliation(s)
- Channabasavaiah B Gurumurthy
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68106-5915, USA.,Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68106-5915, USA
| | - Thomas L Saunders
- Department of Internal Medicine, Division of Genetic Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,Transgenic Animal Model Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Masato Ohtsuka
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine, Isehara, Kanagawa 259-1193, Japan.,The Institute of Medical Sciences, Tokai University, Isehara, Kanagawa 259-1193, Japan
| |
Collapse
|
11
|
Rippe C, Morén B, Liu L, Stenkula KG, Mustaniemi J, Wennström M, Swärd K. NG2/CSPG4, CD146/MCAM and VAP1/AOC3 are regulated by myocardin-related transcription factors in smooth muscle cells. Sci Rep 2021; 11:5955. [PMID: 33727640 PMCID: PMC7966398 DOI: 10.1038/s41598-021-85335-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 02/26/2021] [Indexed: 12/28/2022] Open
Abstract
The present work addressed the hypothesis that NG2/CSPG4, CD146/MCAM, and VAP1/AOC3 are target genes of myocardin-related transcription factors (MRTFs: myocardin/MYOCD, MRTF-A/MKL1, MRTF-B/MKL2) and serum response factor (SRF). Using a bioinformatics approach, we found that CSPG4, MCAM, and AOC3 correlate with MYOCD, MRTF-A/MKL1, and SRF across human tissues. No other transcription factor correlated as strongly with these transcripts as SRF. Overexpression of MRTFs increased both mRNA and protein levels of CSPG4, MCAM, and AOC3 in cultured human smooth muscle cells (SMCs). Imaging confirmed increased staining for CSPG4, MCAM, and AOC3 in MRTF-A/MKL1-transduced cells. MRTFs exert their effects through SRF, and the MCAM and AOC3 gene loci contained binding sites for SRF. SRF silencing reduced the transcript levels of these genes, and time-courses of induction paralleled the direct target ACTA2. MRTF-A/MKL1 increased the activity of promoter reporters for MCAM and AOC3, and transcriptional activation further depended on the chromatin remodeling enzyme KDM3A. CSPG4, MCAM, and AOC3 responded to the MRTF-SRF inhibitor CCG-1423, to actin dynamics, and to ternary complex factors. Coincidental detection of these proteins should reflect MRTF-SRF activity, and beyond SMCs, we observed co-expression of CD146/MCAM, NG2/CSPG4, and VAP1/AOC3 in pericytes and endothelial cells in the human brain. This work identifies highly responsive vascular target genes of MRTF-SRF signaling that are regulated via a mechanism involving KDM3A.
Collapse
Affiliation(s)
- Catarina Rippe
- Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden
| | - Björn Morén
- Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden
| | - Li Liu
- Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden.,Department of Urology, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Karin G Stenkula
- Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden
| | - Johan Mustaniemi
- Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden
| | - Malin Wennström
- Department of Clinical Sciences, Malmö, Lund University, 221 84, Lund, Sweden
| | - Karl Swärd
- Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden.
| |
Collapse
|
12
|
Dhagia V, Kitagawa A, Jacob C, Zheng C, D'Alessandro A, Edwards JG, Rocic P, Gupte R, Gupte SA. G6PD activity contributes to the regulation of histone acetylation and gene expression in smooth muscle cells and to the pathogenesis of vascular diseases. Am J Physiol Heart Circ Physiol 2021; 320:H999-H1016. [PMID: 33416454 PMCID: PMC7988761 DOI: 10.1152/ajpheart.00488.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 11/18/2020] [Accepted: 01/04/2021] [Indexed: 02/05/2023]
Abstract
We aimed to determine 1) the mechanism(s) that enables glucose-6-phosphate dehydrogenase (G6PD) to regulate serum response factor (SRF)- and myocardin (MYOCD)-driven smooth muscle cell (SMC)-restricted gene expression, a process that aids in the differentiation of SMCs, and 2) whether G6PD-mediated metabolic reprogramming contributes to the pathogenesis of vascular diseases in metabolic syndrome (MetS). Inhibition of G6PD activity increased (>30%) expression of SMC-restricted genes and concurrently decreased (40%) the growth of human and rat SMCs ex vivo. Expression of SMC-restricted genes decreased (>100-fold) across successive passages in primary cultures of SMCs isolated from mouse aorta. G6PD inhibition increased Myh11 (47%) while decreasing (>50%) Sca-1, a stem cell marker, in cells passaged seven times. Similarly, CRISPR-Cas9-mediated expression of the loss-of-function Mediterranean variant of G6PD (S188F; G6PDS188F) in rats promoted transcription of SMC-restricted genes. G6PD knockdown or inhibition decreased (48.5%) histone deacetylase (HDAC) activity, enriched (by 3-fold) H3K27ac on the Myocd promoter, and increased Myocd and Myh11 expression. Interestingly, G6PD activity was significantly higher in aortas from JCR rats with MetS than control Sprague-Dawley (SD) rats. Treating JCR rats with epiandrosterone (30 mg/kg/day), a G6PD inhibitor, increased expression of SMC-restricted genes, suppressed Serpine1 and Epha4, and reduced blood pressure. Moreover, feeding SD control (littermates) and G6PDS188F rats a high-fat diet for 4 mo increased Serpine1 and Epha4 expression and mean arterial pressure in SD but not G6PDS188F rats. Our findings demonstrate that G6PD downregulates transcription of SMC-restricted genes through HDAC-dependent deacetylation and potentially augments the severity of vascular diseases associated with MetS.NEW & NOTEWORTHY This study gives detailed mechanistic insight about the regulation of smooth muscle cell (SMC) phenotype by metabolic reprogramming and glucose-6-phosphate dehydrogenase (G6PD) in diabetes and metabolic syndrome. We demonstrate that G6PD controls the chromatin modifications by regulating histone deacetylase (HDAC) activity, which deacetylates histone 3-lysine 9 and 27. Notably, inhibition of G6PD decreases HDAC activity and enriches H3K27ac on myocardin gene promoter to enhance the expression of SMC-restricted genes. Also, we demonstrate for the first time that G6PD inhibitor treatment accentuates metabolic and transcriptomic reprogramming to reduce neointimal formation in coronary artery and large artery elastance in metabolic syndrome rats.
Collapse
MESH Headings
- Acetylation
- Animals
- Cell Line
- Disease Models, Animal
- Female
- Gene Expression Regulation
- Glucosephosphate Dehydrogenase/genetics
- Glucosephosphate Dehydrogenase/metabolism
- Hemodynamics
- Histones/metabolism
- Humans
- Male
- Metabolic Syndrome/enzymology
- Metabolic Syndrome/genetics
- Metabolic Syndrome/pathology
- Metabolic Syndrome/physiopathology
- Mice, Transgenic
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Mutation
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Myosin Heavy Chains/genetics
- Myosin Heavy Chains/metabolism
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Protein Processing, Post-Translational
- Rats, Sprague-Dawley
- Serum Response Factor/genetics
- Serum Response Factor/metabolism
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Vascular Remodeling
- Rats
Collapse
Affiliation(s)
- Vidhi Dhagia
- Department of Pharmacology, New York Medical College, Valhalla, New York
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Atsushi Kitagawa
- Department of Pharmacology, New York Medical College, Valhalla, New York
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Christina Jacob
- Department of Pharmacology, New York Medical College, Valhalla, New York
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Connie Zheng
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - John G Edwards
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Petra Rocic
- Department of Pharmacology, New York Medical College, Valhalla, New York
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Rakhee Gupte
- Raadysan Biotech., Incorporated, Fishkill, New York
| | - Sachin A Gupte
- Department of Pharmacology, New York Medical College, Valhalla, New York
- Department of Physiology, New York Medical College, Valhalla, New York
| |
Collapse
|
13
|
Xu Y, Liang C, Luo Y, Zhang T. MBNL1 regulates isoproterenol-induced myocardial remodelling in vitro and in vivo. J Cell Mol Med 2021; 25:1100-1115. [PMID: 33295096 PMCID: PMC7812249 DOI: 10.1111/jcmm.16177] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/15/2020] [Accepted: 11/25/2020] [Indexed: 12/21/2022] Open
Abstract
Myocardial remodelling is a common phenomenon in cardiovascular diseases, which threaten human health and the quality of life. Due to the lack of effective early diagnosis and treatment methods, the molecular mechanism of myocardial remodelling should be explored in depth. In this study, we observed the high expression of MBNL1 in cardiac tissue and peripheral blood of an isoproterenol (ISO)-induced cardiac hypertrophy mouse model. MBNL1 promoted ISO-induced cardiac hypertrophy and fibrosis by stabilizing Myocardin mRNA in vivo and in vitro. Meanwhile, an increase in MBNL1 may induce the apoptosis of cardiomyocytes treated with ISO via TNF-α signalling. Interestingly, MBNL1 can be activated by p300 in cardiomyocytes treated with ISO. At last, Myocardin can reverse activate the expression of MBNL1. These results suggest that MBNL1 may be a potential target for the early diagnosis and clinical treatment of myocardial remodelling.
Collapse
Affiliation(s)
- Yao Xu
- College of Life Sciences and HealthWuhan University of Science and TechnologyWuhanChina
| | - Chen Liang
- College of Life Sciences and HealthWuhan University of Science and TechnologyWuhanChina
| | - Ying Luo
- College of Biological Science and TechnologyHubei Minzu UniversityEnshiChina
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic diseasesHubei Minzu UniversityEnshiChina
| | - Tongcun Zhang
- College of Life Sciences and HealthWuhan University of Science and TechnologyWuhanChina
| |
Collapse
|
14
|
Burdine RD, Preston CC, Leonard RJ, Bradley TA, Faustino RS. Nucleoporins in cardiovascular disease. J Mol Cell Cardiol 2020; 141:43-52. [PMID: 32209327 DOI: 10.1016/j.yjmcc.2020.02.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/19/2020] [Accepted: 02/25/2020] [Indexed: 01/01/2023]
Abstract
Cardiovascular disease is a pressing health problem with significant global health, societal, and financial burdens. Understanding the molecular basis of polygenic cardiac pathology is thus essential to devising novel approaches for management and treatment. Recent identification of uncharacterized regulatory functions for a class of nuclear envelope proteins called nucleoporins offers the opportunity to understand novel putative mechanisms of cardiac disease development and progression. Consistent reports of nucleoporin deregulation associated with ischemic and dilated cardiomyopathies, arrhythmias and valvular disorders suggests that nucleoporin impairment may be a significant but understudied variable in cardiopathologic disorders. This review discusses and converges existing literature regarding nuclear pore complex proteins and their association with cardiac pathologies, and proposes a role for nucleoporins as facilitators of cardiac disease.
Collapse
Affiliation(s)
- Ryan D Burdine
- Genetics and Genomics Group, Sanford Research, 2301 E. 60(th) Street N., Sioux Falls, SD 57104, United States of America; School of Health Sciences, University of South Dakota, 414 E Clark St, Vermillion, SD 57069, United States of America
| | - Claudia C Preston
- Genetics and Genomics Group, Sanford Research, 2301 E. 60(th) Street N., Sioux Falls, SD 57104, United States of America
| | - Riley J Leonard
- Genetics and Genomics Group, Sanford Research, 2301 E. 60(th) Street N., Sioux Falls, SD 57104, United States of America
| | - Tyler A Bradley
- Genetics and Genomics Group, Sanford Research, 2301 E. 60(th) Street N., Sioux Falls, SD 57104, United States of America
| | - Randolph S Faustino
- Genetics and Genomics Group, Sanford Research, 2301 E. 60(th) Street N., Sioux Falls, SD 57104, United States of America; Department of Pediatrics, Sanford School of Medicine of the University of South Dakota, 1400 W. 22(nd) Street, Sioux Falls, SD 57105, United States of America.
| |
Collapse
|
15
|
Wang Y, Hao Y, Zhao Y, Huang Y, Lai D, Du T, Wan X, Zhu Y, Liu Z, Wang Y, Wang N, Zhang P. TRIM28 and TRIM27 are required for expressions of PDGFRβ and contractile phenotypic genes by vascular smooth muscle cells. FASEB J 2020; 34:6271-6283. [PMID: 32162409 DOI: 10.1096/fj.201902828rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 02/20/2020] [Accepted: 02/27/2020] [Indexed: 12/12/2022]
Abstract
Vascular smooth muscle cells (VSMCs) in the normal arterial media continually express contractile phenotypic markers which are reduced dramatically in response to injury. Tripartite motif-containing proteins are a family of scaffold proteins shown to regulate gene silencing, cell growth, and differentiation. We here investigated the biological role of tripartite motif-containing 28 (TRIM28) and tripartite motif-containing 27 (TRIM27) in VSMCs. We observed that siRNA-mediated knockdown of TRIM28 and TRIM27 inhibited platelet-derived growth factor (PDGF)-induced migration in human VSMCs. Both TRIM28 and TRIM27 can regulate serum response element activity and were required for maintaining the contractile gene expression in human VSMCs. At the same time, TRIM28 and TRIM27 knockdown reduced the expression of PDGF receptor-β (PDGFRβ) and the phosphorylation of its downstream signaling components. Immunoprecipitation showed that TRIM28 formed complexes with TRIM27 through its N-terminal RING-B boxes-Coiled-Coil domain. Furthermore, TRIM28 and TRIM27 were shown to be upregulated and mediate the VSMC contractile marker gene and PDGFRβ expression in differentiating human bone marrow mesenchymal stem cells. In conclusion, we identified that TRIM28 and TRIM27 cooperatively maintain the endogenous expression of PDGFRβ and contractile phenotype of human VSMCs.
Collapse
Affiliation(s)
- Yinfang Wang
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Cardiovascular Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yilong Hao
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanyuan Zhao
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yitong Huang
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dongwu Lai
- Department of Cardiovascular Medicine and Vascular Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Du
- Department of Gastrointestinal Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaohong Wan
- Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Yuefeng Zhu
- Department of Cardiovascular Medicine and Vascular Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zongjun Liu
- Department of Cardiovascular Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Nanping Wang
- The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Peng Zhang
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Cardiovascular Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai Putuo Central School of Clinical Medicine, Anhui Medical University, Hefei, China
| |
Collapse
|
16
|
Nagao M, Lyu Q, Zhao Q, Wirka RC, Bagga J, Nguyen T, Cheng P, Kim JB, Pjanic M, Miano JM, Quertermous T. Coronary Disease-Associated Gene TCF21 Inhibits Smooth Muscle Cell Differentiation by Blocking the Myocardin-Serum Response Factor Pathway. Circ Res 2020; 126:517-529. [PMID: 31815603 PMCID: PMC7274203 DOI: 10.1161/circresaha.119.315968] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/06/2019] [Indexed: 12/12/2022]
Abstract
RATIONALE The gene encoding TCF21 (transcription factor 21) has been linked to coronary artery disease risk by human genome-wide association studies in multiple racial ethnic groups. In murine models, Tcf21 is required for phenotypic modulation of smooth muscle cells (SMCs) in atherosclerotic tissues and promotes a fibroblast phenotype in these cells. In humans, TCF21 expression inhibits risk for coronary artery disease. The molecular mechanism by which TCF21 regulates SMC phenotype is not known. OBJECTIVE To better understand how TCF21 affects the SMC phenotype, we sought to investigate the possible mechanisms by which it regulates the lineage determining MYOCD (myocardin)-SRF (serum response factor) pathway. METHODS AND RESULTS Modulation of TCF21 expression in human coronary artery SMC revealed that TCF21 suppresses a broad range of SMC markers, as well as key SMC transcription factors MYOCD and SRF, at the RNA and protein level. We conducted chromatin immunoprecipitation-sequencing to map SRF-binding sites in human coronary artery SMC, showing that binding is colocalized in the genome with TCF21, including at a novel enhancer in the SRF gene, and at the MYOCD gene promoter. In vitro genome editing indicated that the SRF enhancer CArG box regulates transcription of the SRF gene, and mutation of this conserved motif in the orthologous mouse SRF enhancer revealed decreased SRF expression in aorta and heart tissues. Direct TCF21 binding and transcriptional inhibition at colocalized sites were established by reporter gene transfection assays. Chromatin immunoprecipitation and protein coimmunoprecipitation studies provided evidence that TCF21 blocks MYOCD and SRF association by direct TCF21-MYOCD interaction. CONCLUSIONS These data indicate that TCF21 antagonizes the MYOCD-SRF pathway through multiple mechanisms, further establishing a role for this coronary artery disease-associated gene in fundamental SMC processes and indicating the importance of smooth muscle response to vascular stress and phenotypic modulation of this cell type in coronary artery disease risk.
Collapse
Affiliation(s)
- Manabu Nagao
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Qing Lyu
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine & Dentistry, 601 Elmwood Ave, Rochester, NY 14624
| | - Quanyi Zhao
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Robert C Wirka
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Joetsaroop Bagga
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Trieu Nguyen
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Paul Cheng
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Juyong Brian Kim
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Milos Pjanic
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Joseph M. Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine & Dentistry, 601 Elmwood Ave, Rochester, NY 14624
| | - Thomas Quertermous
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| |
Collapse
|
17
|
Affiliation(s)
- Yi Xie
- From the Departments of Medicine and Pharmacology, Yale University School of Medicine, New Haven, CT
| | - Kathleen A Martin
- From the Departments of Medicine and Pharmacology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
18
|
Lu HS, Schmidt AM, Hegele RA, Mackman N, Rader DJ, Weber C, Daugherty A. Annual Report on Sex in Preclinical Studies: Arteriosclerosis, Thrombosis, and Vascular Biology Publications in 2018. Arterioscler Thromb Vasc Biol 2019; 40:e1-e9. [PMID: 31869272 DOI: 10.1161/atvbaha.119.313556] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Hong S Lu
- From the Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.S.L., A.D.)
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Langone Medical Center, New York, NY (A.M.S.)
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.A.H.)
| | - Nigel Mackman
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC (N.M.)
| | - Daniel J Rader
- Departments of Medicine and Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (D.J.R.)
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität (LMU) and German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Alan Daugherty
- From the Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.S.L., A.D.)
| |
Collapse
|
19
|
Arévalo-Martínez M, Cidad P, García-Mateo N, Moreno-Estar S, Serna J, Fernández M, Swärd K, Simarro M, de la Fuente MA, López-López JR, Pérez-García MT. Myocardin-Dependent Kv1.5 Channel Expression Prevents Phenotypic Modulation of Human Vessels in Organ Culture. Arterioscler Thromb Vasc Biol 2019; 39:e273-e286. [DOI: 10.1161/atvbaha.119.313492] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective:
We have previously described that changes in the expression of Kv channels associate to phenotypic modulation (PM), so that Kv1.3/Kv1.5 ratio is a landmark of vascular smooth muscle cells phenotype. Moreover, we demonstrated that the Kv1.3 functional expression is relevant for PM in several types of vascular lesions. Here, we explore the efficacy of Kv1.3 inhibition for the prevention of remodeling in human vessels, and the mechanisms linking the switch in Kv1.3 /Kv1.5 ratio to PM.
Approach and Results:
Vascular remodeling was explored using organ culture and primary cultures of vascular smooth muscle cells obtained from human vessels. We studied the effects of Kv1.3 inhibition on serum-induced remodeling, as well as the impact of viral vector-mediated overexpression of Kv channels or myocardin knock-down. Kv1.3 blockade prevented remodeling by inhibiting proliferation, migration, and extracellular matrix secretion. PM activated Kv1.3 via downregulation of Kv1.5. Hence, both Kv1.3 blockers and Kv1.5 overexpression inhibited remodeling in a nonadditive fashion. Finally, myocardin knock-down induced vessel remodeling and Kv1.5 downregulation and myocardin overexpression increased Kv1.5, while Kv1.5 overexpression inhibited PM without changing myocardin expression.
Conclusions:
We demonstrate that Kv1.5 channel gene is a myocardin-regulated, vascular smooth muscle cells contractile marker. Kv1.5 downregulation upon PM leaves Kv1.3 as the dominant Kv1 channel expressed in dedifferentiated cells. We demonstrated that the inhibition of Kv1.3 channel function with selective blockers or by preventing Kv1.5 downregulation can represent an effective, novel strategy for the prevention of intimal hyperplasia and restenosis of the human vessels used for coronary angioplasty procedures.
Collapse
Affiliation(s)
- Marycarmen Arévalo-Martínez
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
| | - Pilar Cidad
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
| | - Nadia García-Mateo
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
| | - Sara Moreno-Estar
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
| | - Julia Serna
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
| | - Mirella Fernández
- Cardiovascular Surgery Department, Hospital Clínico Universitario de Valladolid, Spain (M.F.)
| | - Karl Swärd
- Department of Experimental Medical Science, University of Lund, Sweden (K.S.)
| | - María Simarro
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
- Departamento de Enfermería, Universidad de Valladolid, Spain (M.S.)
| | - Miguel A. de la Fuente
- Instituto de Biología y Genética Molecular (IBGM), CSIC, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., M.S., M.A.d.l.F.)
- Departamento de Biología Celular, Universidad de Valladolid, Spain (M.A.d.l.F.)
| | - José R. López-López
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
| | - M. Teresa Pérez-García
- From the Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid, Spain (M.A.-M., P.C., N.G.-M., S.M.-E., J.S., J.R.L.-L., M.T.P.-G.)
| |
Collapse
|
20
|
Li F, Shi J, Lu HS, Zhang H. Functional Genomics and CRISPR Applied to Cardiovascular Research and Medicine. Arterioscler Thromb Vasc Biol 2019; 39:e188-e194. [PMID: 31433696 DOI: 10.1161/atvbaha.119.312579] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Fang Li
- From the Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York (F.L., J.S., H.Z.)
| | - Jianting Shi
- From the Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York (F.L., J.S., H.Z.)
| | - Hong S Lu
- Department of Physiology, Saha Cardiovascular Research Center, University of Kentucky, Lexington (H.S.L.)
| | - Hanrui Zhang
- From the Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York (F.L., J.S., H.Z.)
| |
Collapse
|
21
|
Transcription factor TEAD1 is essential for vascular development by promoting vascular smooth muscle differentiation. Cell Death Differ 2019; 26:2790-2806. [PMID: 31024075 DOI: 10.1038/s41418-019-0335-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 03/04/2019] [Accepted: 04/04/2019] [Indexed: 12/25/2022] Open
Abstract
TEAD1 (TEA domain transcription factor 1), a transcription factor known for the functional output of Hippo signaling, is important for tumorigenesis. However, the role of TEAD1 in the development of vascular smooth muscle cell (VSMC) is unknown. To investigate cell-specific role of Tead1, we generated cardiomyocyte (CMC) and VSMC-specific Tead1 knockout mice. We found CMC/VSMC-specific deletion of Tead1 led to embryonic lethality by E14.5 in mice due to hypoplastic cardiac and vascular walls, as a result of impaired CMC and VSMC proliferation. Whole transcriptome analysis revealed that deletion of Tead1 in CMCs/VSMCs downregulated expression of muscle contractile genes and key transcription factors including Pitx2c and myocardin. In vitro studies demonstrated that PITX2c and myocardin rescued TEAD1-dependent defects in VSMC differentiation. We further identified Pitx2c as a novel transcriptional target of TEAD1, and PITX2c exhibited functional synergy with myocardin by directly interacting with myocardin, leading to augment the differentiation of VSMC. In summary, our study reveals a critical role of Tead1 in cardiovascular development in mice, but also identifies a novel regulatory mechanism, whereby Tead1 functions upstream of the genetic regulatory hierarchy for establishing smooth muscle contractile phenotype.
Collapse
|
22
|
Mittal A, Rana S, Sharma R, Kumar A, Prasad R, Raut SK, Sarkar S, Saikia UN, Bahl A, Dhandapany PS, Khullar M. Myocardin ablation in a cardiac-renal rat model. Sci Rep 2019; 9:5872. [PMID: 30971740 PMCID: PMC6458122 DOI: 10.1038/s41598-019-42009-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 03/13/2019] [Indexed: 11/09/2022] Open
Abstract
Cardiorenal syndrome is defined by primary heart failure conditions influencing or leading to renal injury or dysfunction. Dilated cardiomyopathy (DCM) is a major co-existing form of heart failure (HF) with renal diseases. Myocardin (MYOCD), a cardiac-specific co-activator of serum response factor (SRF), is increased in DCM porcine and patient cardiac tissues and plays a crucial role in the pathophysiology of DCM. Inhibiting the increased MYOCD has shown to be partially rescuing the DCM phenotype in porcine model. However, expression levels of MYOCD in the cardiac tissues of the cardiorenal syndromic patients and the effect of inhibiting MYOCD in a cardiorenal syndrome model remains to be explored. Here, we analyzed the expression levels of MYOCD in the DCM patients with and without renal diseases. We also explored, whether cardiac specific silencing of MYOCD expression could ameliorate the cardiac remodeling and improve cardiac function in a renal artery ligated rat model (RAL). We observed an increase in MYOCD levels in the endomyocardial biopsies of DCM patients associated with renal failure compared to DCM alone. Silencing of MYOCD in RAL rats by a cardiac homing peptide conjugated MYOCD siRNA resulted in attenuation of cardiac hypertrophy, fibrosis and restoration of the left ventricular functions. Our data suggest hyper-activation of MYOCD in the pathogenesis of the cardiorenal failure cases. Also, MYOCD silencing showed beneficial effects by rescuing cardiac hypertrophy, fibrosis, size and function in a cardiorenal rat model.
Collapse
Affiliation(s)
- Anupam Mittal
- Centre for Cardiovascular Biology and Disease, Institute for Stem Cell Biology and Regenerative Medicine (inStem), Bangalore, India.,Department of Cardiology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Santanu Rana
- Department of Zoology, University of Calcutta, Kolkata, India
| | - Rajni Sharma
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Akhilesh Kumar
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Rishikesh Prasad
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Satish K Raut
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Uma Nahar Saikia
- Department of Histopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ajay Bahl
- Department of Cardiology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Perundurai S Dhandapany
- Centre for Cardiovascular Biology and Disease, Institute for Stem Cell Biology and Regenerative Medicine (inStem), Bangalore, India. .,The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA. .,Department of Medicine, Oregon Health and Science University, Portland, OR, USA. .,Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA.
| | - Madhu Khullar
- Department of Experimental Medicine and Biotechnology, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
23
|
Herring BP, Hoggatt AM, Gupta A, Wo JM. Gastroparesis is associated with decreased FOXF1 and FOXF2 in humans, and loss of FOXF1 and FOXF2 results in gastroparesis in mice. Neurogastroenterol Motil 2019; 31:e13528. [PMID: 30565344 PMCID: PMC6821388 DOI: 10.1111/nmo.13528] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/02/2018] [Accepted: 11/12/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS The transcription factors FOXF1 and FOXF2 have been implicated in the development of the gastrointestinal tract but their role in adults or in gastrointestinal diseases is poorly understood. We have recently shown that expression of serum response factor (SRF), a transcription factor whose activity is modulated by FOXF proteins, is decreased in the stomach muscularis of patients with gastroparesis. The aim of the current study was to determine whether FOXF expression is decreased in gastroparesis patients and whether loss of FOXF1 and/or FOXF2 from adult smooth muscle is sufficient to impair gastric emptying in mice. METHODS Full-thickness stomach biopsy samples were collected from control subjects and from patients with gastroparesis. mRNA was isolated from the muscularis externa, and FOXF mRNA expression levels were determined by quantitative reverse transcriptase (RT)-PCR. Foxf1 and Foxf2 were knocked out together and separately from smooth muscle cells in adult mice, and the subsequent effect on liquid gastric emptying and contractile protein expression was determined. KEY RESULTS Expression of FOXF1 and FOXF2 is decreased in smooth muscle tissue from gastroparesis patients. Knockout of Foxf1 and Foxf2 together, but not alone, from mouse smooth muscle resulted in delayed liquid gastric emptying. Foxf1/2 double knockout mice had decreased expression of smooth muscle contractile proteins, SRF, and myocardin in stomach muscularis. CONCLUSIONS AND INFERENCES Our findings suggest that decreased expression of FOXF1 and FOXF2 may be contributing to the impaired gastric emptying seen in gastroparesis patients.
Collapse
Affiliation(s)
- B. Paul Herring
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202.,Correspondence: Paul Herring, Department of Cellular and Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis IN, 46202, Phone: (317) 278-1785, FAX: (317) 274-3318,
| | - April M. Hoggatt
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Anita Gupta
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - John M. Wo
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
24
|
Miano JM, Long X. CRISPR-tagging mice in aging research. Aging (Albany NY) 2018; 10:2226-2227. [PMID: 30243018 PMCID: PMC6188475 DOI: 10.18632/aging.101566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 09/18/2018] [Indexed: 11/25/2022]
Affiliation(s)
- Joseph M Miano
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Xiaochun Long
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| |
Collapse
|
25
|
Gao P, Wu W, Ye J, Lu YW, Adam AP, Singer HA, Long X. Transforming growth factor β1 suppresses proinflammatory gene program independent of its regulation on vascular smooth muscle differentiation and autophagy. Cell Signal 2018; 50:160-170. [PMID: 30006123 DOI: 10.1016/j.cellsig.2018.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/19/2018] [Accepted: 07/09/2018] [Indexed: 01/01/2023]
Abstract
Transforming growth factor β (TGFβ) signaling plays crucial roles in maintaining vascular integrity and homeostasis, and is established as a strong activator of vascular smooth muscle cell (VSMC) differentiation. Chronic inflammation is a hallmark of various vascular diseases. Although TGFβ signaling has been suggested to be protective against inflammatory aortic aneurysm progression, its exact effects on VSMC inflammatory process and the underlying mechanisms are not fully unraveled. Here we revealed that TGFβ1 suppressed the expression of a broad array of proinflammatory genes while potently induced the expression of contractile genes in cultured primary human coronary artery SMCs (HCASMCs). The regulation of TGFβ1 on VSMC contractile and proinflammatory gene programs appeared to occur in parallel and both processes were through a SMAD4-dependent canonical pathway. We also showed evidence that the suppression of TGFβ1 on VSMC proinflammatory genes was mediated, at least partially through the blockade of signal transducer and activator of transcription 3 (STAT3) and NF-κB pathways. Interestingly, our RNA-seq data also revealed that TGFβ1 suppressed gene expression of a battery of autophagy mediators, which was validated by western blot for the conversion of microtubule-associated protein light chain 3 (LC3) and by immunofluo-rescence staining for LC3 puncta. However, impairment of VSMC autophagy by ATG5 deletion failed to rescue TGFβ1 influence on both VSMC contractile and proinflammatory gene programs, suggesting that TGFβ1-regulated VSMC differentiation and inflammation are not attributed to TGFβ1 suppression on autophagy. In summary, our results demonstrated an important role of TGFβ signaling in suppressing proinflammatory gene program in cultured primary human VSMCs via the blockade on STAT3 and NF-κB pathway, therefore providing novel insights into the mechanisms underlying the protective role of TGFβ signaling in vascular diseases.
Collapse
Affiliation(s)
- Ping Gao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Wen Wu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Jiemei Ye
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Yao Wei Lu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Alejandro Pablo Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States; Department of Ophthalmology, Albany Medical College, Albany, NY, United States
| | - Harold A Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Xiaochun Long
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States.
| |
Collapse
|