51
|
张 涛, 李 维, 邱 晓, 刘 百, 李 高, 冯 才, 廖 俊, 林 康. [CRISPR/Cas9-mediated TEAD1 knockout induces phenotypic modulation of corpus cavernosum smooth muscle cells in diabetic rats with erectile dysfunction]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:567-573. [PMID: 33963717 PMCID: PMC8110442 DOI: 10.12122/j.issn.1673-4254.2021.04.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To construct a corpus cavemosum smooth muscle cell (CCSMCs) line with TEAD1 knockout from diabetic rats with erectile dysfunction (ED) using CRISPR/Cas9 technology and explore the role of TEAD1 in phenotypic modulation of CCSMCs in diabetic rats with ED. OBJECTIVE Models of diabetic ED were established in male Sprague-Dawley rats by intraperitoneal injection of streptozotocin. CCSMCs from the rat models were primarily cultured and identified with immunofluorescence assay. Three sgRNAs (sgRNA-1, sgRNA-2 and sgRNA-3) were transfected via lentiviral vectors into 293T cells to prepare the sgRNA-Cas9 lentivirus. CCSMCs from diabetic rats with ED were infected by the lentivirus, and the cellular expression of TEAD1 protein was detected using Western blotting. In CCSMCs infected with the sgRNA-Cas9 lentivirus (CCSMCs-sgRNA-2), or the empty lentiviral vector (CCSMCs-sgRNA-NC) and the blank control cells (CCSMCs-CK), the expressions of cellular phenotypic markers SMMHC, calponin and PCNA at the mRNA and protein levels were detected using real-time fluorescence quantitative RT-PCR (qRT-PCR) and Western blotting, respectively. OBJECTIVE The primarily cultured CCSMCs from diabetic rats with ED showed a high α-SMA-positive rate of over 95%. The recombinant lentivirus of TEAD1-sgRNA was successfully packaged, and stable TEAD1-deficient CCSMC lines derived from diabetic rat with ED were obtained. Western blotting confirmed that the protein expression of TEAD1 in TEAD1-sgRNA-2 group was the lowest (P < 0.05), and this cell line was used in subsequent experiment. The results of qRT-PCR and Western blotting showed significantly up-regulated expressions of SMMHC and calponin (all P < 0.05) and down-regulated expression of PCNA (all P < 0.05) at both the mRNA and protein levels in TEAD1-deficient CCSMCs from diabetic rats with ED. OBJECTIVE We successfully constructed a stable CCSMCs line with CRISPR/Cas9-mediated TEAD1 knockout from diabetic rats with ED. TEAD1 gene knockout can induce phenotype transformation of the CCSMCs from diabetic rats with ED from the synthetic to the contractile type.
Collapse
Affiliation(s)
- 涛 张
- 广东省第二人民医院泌尿外科,广东 广州 510317Department of Urology, Second Guangdong Provincial People's Hospital, Guangzhou 510317, China
| | - 维丽 李
- 南方医科大学南方医院妇产科,广东 广州 510515Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 晓拂 邱
- 广东省第二人民医院泌尿外科,广东 广州 510317Department of Urology, Second Guangdong Provincial People's Hospital, Guangzhou 510317, China
| | - 百川 刘
- 广东省第二人民医院泌尿外科,广东 广州 510317Department of Urology, Second Guangdong Provincial People's Hospital, Guangzhou 510317, China
| | - 高远 李
- 广东省第二人民医院泌尿外科,广东 广州 510317Department of Urology, Second Guangdong Provincial People's Hospital, Guangzhou 510317, China
| | - 才鑫 冯
- 广东省第二人民医院泌尿外科,广东 广州 510317Department of Urology, Second Guangdong Provincial People's Hospital, Guangzhou 510317, China
| | - 俊发 廖
- 广东省第二人民医院泌尿外科,广东 广州 510317Department of Urology, Second Guangdong Provincial People's Hospital, Guangzhou 510317, China
| | - 康健 林
- 广东省第二人民医院泌尿外科,广东 广州 510317Department of Urology, Second Guangdong Provincial People's Hospital, Guangzhou 510317, China
| |
Collapse
|
52
|
Wen H, Shi W, Ge S, Li J, Zuo L, Liu M. [Value of prediction models for prognosis prediction of colorectal cancer: an analysis based on TCPA database]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:439-446. [PMID: 33849837 DOI: 10.12122/j.issn.1673-4254.2021.03.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To assess the value of the combination of multiple proteins in predicting the prognosis of colorectal cancer (CRC) through bioinformatics analysis. OBJECTIVE The protein expression and clinical data were downloaded from TCPA database. Perl and R were used to screen the prognostic-related proteins, and through Cox analysis, the proteins that served as independent prognostic factors of CRC were identified to build the prediction model. Survival analyses were conducted for each of the proteins included in the prediction model and the risk score of the model, and risk curves was drawn for the risk score and the patients' survival status to verify the performance of the model. Independent prognosis analysis and ROC analysis were used to assess the value and advantages of the model in prognosis prediction. The interactions between the proteins included in the model and the differential expressions of the key genes related with the proteins were analyzed. OBJECTIVE Six proteins were screened for model construction. Compared with a single gene, the model showed much greater prognostic value for CRC. Independent prognostic analysis showed that the risk score of the prediction model was significantly related with the prognosis (P < 0.001), and the model could be used as an independent risk factor for prognostic assessment of the patients. ROC analysis showed that the model had good specificity and sensitivity for prognostic prediction (AUC=0.734). Protein interactions showed that BID, SLC1A5 and SRC_pY527 were significantly correlated with other proteins (P < 0.001), and SLC1A5 and SRC_pY527 had the most significant interactions with other proteins (P < 0.001). Except for those of INPP4B, the key genes related with the proteins in the prediction model had significant differential expressions at the mRNA level in CRC (P < 0.05). OBJECTIVE The prediction model constructed based on 6 proteins has good prognostic value for CRC. The proteins SLC1A5 and SRC_pY527 play key roles in the prognosis of CRC, and SRC_pY527 may regulate the occurrence and progression of CRC through the SRC/AKT/MAPK signal axis and thus may serve as a new therapeutic target of CRC.
Collapse
Affiliation(s)
- H Wen
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - W Shi
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - S Ge
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - J Li
- Department of Laboratory Medicine, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - L Zuo
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - M Liu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| |
Collapse
|
53
|
Osman I, Dong K, Kang X, Yu L, Xu F, Ahmed ASI, He X, Shen J, Hu G, Zhang W, Zhou J. YAP1/TEAD1 upregulate platelet-derived growth factor receptor beta to promote vascular smooth muscle cell proliferation and neointima formation. J Mol Cell Cardiol 2021; 156:20-32. [PMID: 33753119 DOI: 10.1016/j.yjmcc.2021.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/22/2021] [Accepted: 03/13/2021] [Indexed: 12/19/2022]
Abstract
We have previously demonstrated that the transcription co-factor yes-associated protein 1 (YAP1) promotes vascular smooth muscle cell (VSMC) de-differentiation. Yet, the role and underlying mechanisms of YAP1 in neointima formation in vivo remain unclear. The goal of this study was to investigate the role of VSMC-expressed YAP1 in vascular injury-induced VSMC proliferation and delineate the mechanisms underlying its action. Experiments employing gain- or loss-of-function of YAP1 demonstrated that YAP1 promotes human VSMC proliferation. Mechanistically, we identified platelet-derived growth factor receptor beta (PDGFRB) as a novel YAP1 target gene that confers the YAP1-dependent hyper-proliferative effects in VSMCs. Furthermore, we identified TEA domain transcription factor 1 (TEAD1) as a key transcription factor that mediates YAP1-dependent PDGFRβ expression. ChIP assays demonstrated that TEAD1 is enriched at a PDGFRB gene enhancer. Luciferase reporter assays further demonstrated that YAP1 and TEAD1 co-operatively activate the PDGFRB enhancer. Consistent with these observations, we found that YAP1 expression is upregulated after arterial injury and correlates with PDGFRβ expression and VSMC proliferation in vivo. Using a novel inducible SM-specific Yap1 knockout mouse model, we found that the specific deletion of Yap1 in adult VSMCs is sufficient to attenuate arterial injury-induced neointima formation, largely due to inhibited PDGFRβ expression and VSMC proliferation. Our study unravels a novel mechanism by which YAP1/TEAD1 promote VSMC proliferation via transcriptional induction of PDGFRβ, thereby enhancing PDGF-BB downstream signaling and promoting neointima formation.
Collapse
Affiliation(s)
- Islam Osman
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Kunzhe Dong
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Xiuhua Kang
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Luyi Yu
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Fei Xu
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Abu Shufian Ishtiaq Ahmed
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Xiangqin He
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Jian Shen
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Guoqing Hu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Wei Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Jiliang Zhou
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States.
| |
Collapse
|
54
|
Zhang CY, Hu YC, Zhang Y, Ma WD, Song YF, Quan XH, Guo X, Wang CX. Glutamine switches vascular smooth muscle cells to synthetic phenotype through inhibiting miR-143 expression and upregulating THY1 expression. Life Sci 2021; 277:119365. [PMID: 33741416 DOI: 10.1016/j.lfs.2021.119365] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/03/2021] [Accepted: 03/07/2021] [Indexed: 11/27/2022]
Abstract
AIMS Vascular smooth muscle cells (VSMCs) are involved in the pathogenesis of many human cardiovascular diseases. They modulate their phenotype from "contractile" to "synthetic" in response to changes in local environmental cues. How glutamine regulates the differentiation of VSMCs and the underlying mechanisms remain largely unknown. MAIN METHODS Here, we explored the effects of various doses of glutamine (0 mM, 1 mM, 2 mM, and 4 mM) on the proliferation, migration, and phenotypic switch of human VSMCs in vitro. Glutamine dose-dependently enhanced VSMC proliferation, and markedly increased VSMC migration. KEY FINDINGS Notably, glutamine promoted the phenotypic switch of VSMCs towards a synthetic phenotype, as evidenced by significantly decreased expression of contractile markers myosin heavy chain 11 (MYH11) and calponin while increased expression of synthetic markers collagen I and vimentin. Importantly, these changes upon glutamine treatments were attenuated after additional treatments with glutamine metabolism inhibitor BPTES. Additionally, glutamine downregulated miR-143 expression, and miR-143 inactivation alone resulted in enhanced proliferation, migration, and promoted the synthetic phenotype of VSMCs. Moreover, Thy-1 cell surface antigen (THY1) was validated as a downstream target of miR-143, and THY1 expression was upregulated by glutamine in VSMCs. Furthermore, either miR-143 overexpression or THY1 silencing abolished the effect of glutamine on proliferation, migration, and phenotypic switch of VSMCs, supporting a novel glutamine-miR-143-THY1 pathway in modulating VSMC functions. SIGNIFICANCE This study demonstrated a novel mechanism of glutamine in modulation of VSMC phenotypic switch by targeting miR-143 and THY1, and provides significant insight on targeted therapy of patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Chun-Yan Zhang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, 710004 Xi'an, China
| | - Yan-Chao Hu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, 710004 Xi'an, China
| | - Yan Zhang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, 710004 Xi'an, China
| | - Wei-Dong Ma
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, 710004 Xi'an, China
| | - Ya-Fan Song
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, 710004 Xi'an, China
| | - Xiao-Hui Quan
- Department of Cardiovascular Medicine, Xi'an No.1 Hospital, 30 Fen Xiang, South Street, 710004 Xi'an, China
| | - Xuan Guo
- Department of Cardiovascular Medicine, Xi'an No.1 Hospital, 30 Fen Xiang, South Street, 710004 Xi'an, China
| | - Cong-Xia Wang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, 710004 Xi'an, China.
| |
Collapse
|
55
|
Manno G, Filorizzo C, Fanale D, Brando C, Di Lisi D, Lunetta M, Bazan V, Russo A, Novo G. Role of the HIPPO pathway as potential key player in the cross talk between oncology and cardiology. Crit Rev Oncol Hematol 2021; 159:103246. [PMID: 33545354 DOI: 10.1016/j.critrevonc.2021.103246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/19/2022] Open
Abstract
The HIPPO pathway (HP) is a highly conserved kinase cascade that affects organ size by regulating proliferation, cell survival and differentiation. Discovered in Drosophila melanogaster to early 2000, it immediately opened wide frontiers in the field of research. Over the last years the field of knowledge on HP is quickly expanding and it is thought will offer many answers on complex pathologies. Here, we summarized the results of several studies that have investigated HP signaling both in oncology than in cardiology field, with an overview on future perspectives in cardiology research.
Collapse
Affiliation(s)
- Girolamo Manno
- Cardiology Unit, University Hospital P. Giaccone, Department of Excellence of Sciences for Health Promotion and Mothernal-Child Care, Internal Medicine and Specialities (ProMISE) "G. D'Alessandro", Palermo, Italy
| | - Clarissa Filorizzo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Italy
| | - Daniele Fanale
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Italy
| | - Chiara Brando
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Italy
| | - Daniela Di Lisi
- Cardiology Unit, University Hospital P. Giaccone, Department of Excellence of Sciences for Health Promotion and Mothernal-Child Care, Internal Medicine and Specialities (ProMISE) "G. D'Alessandro", Palermo, Italy
| | - Monica Lunetta
- Cardiology Unit, University Hospital P. Giaccone, Department of Excellence of Sciences for Health Promotion and Mothernal-Child Care, Internal Medicine and Specialities (ProMISE) "G. D'Alessandro", Palermo, Italy
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Section of Medical Oncology, Palermo, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Italy.
| | - Giuseppina Novo
- Cardiology Unit, University Hospital P. Giaccone, Department of Excellence of Sciences for Health Promotion and Mothernal-Child Care, Internal Medicine and Specialities (ProMISE) "G. D'Alessandro", Palermo, Italy
| |
Collapse
|
56
|
Shi J, Yang Y, Cheng A, Xu G, He F. Metabolism of vascular smooth muscle cells in vascular diseases. Am J Physiol Heart Circ Physiol 2020; 319:H613-H631. [PMID: 32762559 DOI: 10.1152/ajpheart.00220.2020] [Citation(s) in RCA: 200] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Vascular smooth muscle cells (VSMCs) are the fundamental component of the medial layer of arteries and are essential for arterial physiology and pathology. It is becoming increasingly clear that VSMCs can alter their metabolism to fulfill the bioenergetic and biosynthetic requirements. During vascular injury, VSMCs switch from a quiescent "contractile" phenotype to a highly migratory and proliferative "synthetic" phenotype. Recent studies have found that the phenotype switching of VSMCs is driven by a metabolic switch. Metabolic pathways, including aerobic glycolysis, fatty acid oxidation, and amino acid metabolism, have distinct, indispensable roles in normal and dysfunctional vasculature. VSMCs metabolism is also related to the metabolism of endothelial cells. In the present review, we present a brief overview of VSMCs metabolism and how it regulates the progression of several vascular diseases, including atherosclerosis, systemic hypertension, diabetes, pulmonary hypertension, vascular calcification, and aneurysms, and the effect of the risk factors for vascular disease (aging, cigarette smoking, and excessive alcohol drinking) on VSMC metabolism to clarify the role of VSMCs metabolism in the key pathological process.
Collapse
Affiliation(s)
- Jia Shi
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Yang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anying Cheng
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Xu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan He
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
57
|
Heng BC, Zhang X, Aubel D, Bai Y, Li X, Wei Y, Fussenegger M, Deng X. Role of YAP/TAZ in Cell Lineage Fate Determination and Related Signaling Pathways. Front Cell Dev Biol 2020; 8:735. [PMID: 32850847 PMCID: PMC7406690 DOI: 10.3389/fcell.2020.00735] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/15/2020] [Indexed: 12/11/2022] Open
Abstract
The penultimate effectors of the Hippo signaling pathways YAP and TAZ, are transcriptional co-activator proteins that play key roles in many diverse biological processes, ranging from cell proliferation, tumorigenesis, mechanosensing and cell lineage fate determination, to wound healing and regeneration. In this review, we discuss the regulatory mechanisms by which YAP/TAZ control stem/progenitor cell differentiation into the various major lineages that are of interest to tissue engineering and regenerative medicine applications. Of particular interest is the key role of YAP/TAZ in maintaining the delicate balance between quiescence, self-renewal, proliferation and differentiation of endogenous adult stem cells within various tissues/organs during early development, normal homeostasis and regeneration/healing. Finally, we will consider how increasing knowledge of YAP/TAZ signaling might influence the trajectory of future progress in regenerative medicine.
Collapse
Affiliation(s)
- Boon C. Heng
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, China
- Faculty of Science and Technology, Sunway University, Subang Jaya, Malaysia
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, China
| | - Dominique Aubel
- IUTA Department Genie Biologique, Universite Claude Bernard Lyon 1, Villeurbanne, France
| | - Yunyang Bai
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xiaochan Li
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Yan Wei
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH-Zürich, Basel, Switzerland
| | - Xuliang Deng
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, China
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
58
|
Xue M, Hong W, Jiang J, Zhao F, Gao X. Circular RNA circ-LDLRAD3 serves as an oncogene to promote non-small cell lung cancer progression by upregulating SLC1A5 through sponging miR-137. RNA Biol 2020; 17:1811-1822. [PMID: 32658600 DOI: 10.1080/15476286.2020.1789819] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Circular RNAs (circRNAs) are closely associated with the development of non-small cell lung cancer (NSCLC); however, it is still unclear whether circular RNA circ-LDLRAD3 participated in the regulation of NSCLC progression. In this study, we found that circ-LDLRAD3 was high-expressed and miR-137 was low-expressed in NSCLC tissues and cells compared to their normal counterparts, which showed negative correlations in NSCLC tissues. Further experiments validated that miR-137 could be sponged and inhibited by circ-LDLRAD3 in NSCLC cells. In addition, knock-down of circ-LDLRAD3 and miR-137 overexpression promoted NSCLC cell apoptosis, and inhibited cell proliferation and invasion. Similarly, upregulation of circ-LDLRAD3 or miR-137 ablation had opposite effects on the above cell functions. Besides, the glutamine transporter SLC1A5 was validated to be the downstream target of circ-LDLRAD3 and miR-137, and upregulated circ-LDLRAD3 increased SLC1A5 expression levels by downregulating miR-137. Furthermore, the effects of downregulated circ-LDLRAD3 on cell proliferation, apoptosis and mobility were all reversed by knocking down miR-137 and overexpressing SLC1A5. Taken together, this in vitro study found that knock-down of circ-LDLRAD3 inhibited the development of NSCLC by regulating miR-137/SLC1A5 axis.
Collapse
Affiliation(s)
- Min Xue
- Department of Respiratory Medicine, Minhang Hospital, Fudan University , Shanghai, China
| | - Weijun Hong
- Department of Respiratory Medicine, Minhang Hospital, Fudan University , Shanghai, China
| | - Jun Jiang
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Science, Fudan University , Shanghai, China
| | - Fang Zhao
- Department of Laboratory, Minhang Hospital, Fudan University , Shanghai, China
| | - Xiwen Gao
- Department of Respiratory Medicine, Minhang Hospital, Fudan University , Shanghai, China
| |
Collapse
|
59
|
Williams R, The Editors. Circulation Research
“In This Issue” Anthology. Circ Res 2020. [DOI: 10.1161/res.0000000000000406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
60
|
Sun X, Li S, Gan X, Chen K, Yang D, Yang Y. NF2 deficiency accelerates neointima hyperplasia following vascular injury via promoting YAP-TEAD1 interaction in vascular smooth muscle cells. Aging (Albany NY) 2020; 12:9726-9744. [PMID: 32422606 PMCID: PMC7288949 DOI: 10.18632/aging.103240] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 03/31/2020] [Indexed: 01/12/2023]
Abstract
Neurofibromin 2 (NF2), a potent tumor suppressor, is reported to inhibit proliferation in several cell types. The role of NF2 in neointima hyperplasia after vascular injury is unknown. We explored the role of NF2 in proliferation, migration of vascular smooth muscle cell (VSMC) and neointima hyperplasia after vascular injury. NF2 phosphorylation was elevated in VSMC subjected to platelet-derived growth factor (PDGF)-BB and in artery subjected to vascular injury. Mice deficient for Nf2 in VSMC showed enhanced neointima hyperplasia after injury, increased proliferation and migration of VSMC after PDGF-BB treatment. Mechanistically, we observed increased nuclear p-NF2, declined p-Yes-Associated Protein (YAP), nuclear translocation of YAP after PDGF-BB treatment or injury. NF2 knockdown or YAP overexpression showed similar phenotype in VSMC proliferation, migration and neointima hyperplasia. YAP inhibition abolished the above effects mediated by NF2 knockdown. Finally, NF2 knockdown further promoted YAP-TEA Domain Transcription Factor 1 (TEAD1) interaction after PDGF-BB treatment. Inhibition of TEAD1 blocked PDGF-BB-induced VSMC proliferation and migration, which were not reversed by either NF2 knockdown or YAP overexpression. In conclusion, NF2 knockdown promotes VSMC proliferation, migration and neointima hyperplasia after vascular injury via inducing YAP-TEAD1 interaction.
Collapse
Affiliation(s)
- Xiongshan Sun
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Shuang Li
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Xueqing Gan
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Ken Chen
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Dachun Yang
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Yongjian Yang
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu 610083, China
| |
Collapse
|
61
|
Tang J, Wang H, Huang X, Li F, Zhu H, Li Y, He L, Zhang H, Pu W, Liu K, Zhao H, Bentzon JF, Yu Y, Ji Y, Nie Y, Tian X, Zhang L, Gao D, Zhou B. Arterial Sca1 + Vascular Stem Cells Generate De Novo Smooth Muscle for Artery Repair and Regeneration. Cell Stem Cell 2019; 26:81-96.e4. [PMID: 31883835 DOI: 10.1016/j.stem.2019.11.010] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/24/2019] [Accepted: 11/18/2019] [Indexed: 01/09/2023]
Abstract
Rapid regeneration of smooth muscle after vascular injury is essential for maintaining arterial function. The existence and putative roles of resident vascular stem cells (VSCs) in artery repair are controversial, and vessel regeneration is thought to be mediated by proliferative expansion of pre-existing smooth muscle cells (SMCs). Here, we performed cell fate mapping and single-cell RNA sequencing to identify Sca1+ VSCs in the adventitial layer of artery walls. After severe injury, Sca1+ VSCs migrate into the medial layer and generate de novo SMCs, which subsequently expand more efficiently compared with pre-existing smooth muscle. Genetic lineage tracing using dual recombinases distinguished a Sca1+PDGFRa+ VSC subpopulation that generates SMCs, and genetic ablation of Sca1+ VSCs or specific knockout of Yap1 in Sca1+ VSCs significantly impaired artery repair. These findings provide genetic evidence of a bona fide Sca1+ VSC population that produces SMCs and delineates their critical role in vessel repair.
Collapse
Affiliation(s)
- Juan Tang
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Haixiao Wang
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiuzhen Huang
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Fei Li
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Huan Zhu
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan Li
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lingjuan He
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hui Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wenjuan Pu
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kuo Liu
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Huan Zhao
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jacob Fog Bentzon
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain; Deparment of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Ying Yu
- Department of Pharmacology and Tianjin Key Laboratory of Inflammatory Biology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing 211100, China; The Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211100, China
| | - Yu Nie
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xueying Tian
- Key Laboratory of Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Li Zhang
- The Department of Cardiology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Dong Gao
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| | - Bin Zhou
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Key Laboratory of Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou 510632, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
62
|
Fan X, Li Z, Wang X, Wang J, Hao Z. Silencing of KPNA2 inhibits high glucose-induced podocyte injury via inactivation of mTORC1/p70S6K signaling pathway. Biochem Biophys Res Commun 2019; 521:1017-1023. [PMID: 31727365 DOI: 10.1016/j.bbrc.2019.10.200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 10/31/2019] [Indexed: 12/17/2022]
Abstract
Dysregulation of apoptotic and autophagic function are characterized as the main pathogeneses of diabetic nephropathy (DN). It has been reported that Karyopherin Alpha 2 (KPNA2) contributes to apoptosis and autophagy in various cells, but its role in DN development remains unknown. The purpose of present study was to explore the function and underling mechanisms of KPNA2 in development of DN. In this study, 30 mM high glucose (HG)-evoked podocytes were used as DN model. The expression of KPNA2 was detected by qRT-PCR and Western blot assays. The cell viability was tested by CCK-8 kit, the apoptosis was measured using flow cytometry assay, the apoptotic and the autophagy related genes was detected by Western blot. Our results indicated that KPNA2 was significantly increased after HG stimulation. Knockdown of KPNA2 inhibited apoptosis, and promoted cell viability and autophagy in HG-treated podocytes. In addition, silencing of KPNA2 deactivated mTORC1/p70S6K pathway activation via regulating SLC1A5. Further results demonstrated that activating mTORC1/p70S6K pathway strongly ameliorated the effect of KPNA2 on cell viability, apoptosis and autophagy. Therefore, our study suggested that knockdown of KPNA2 rescued HG-induced injury via blocking activation of mTORC1/p70S6K pathway by mediating SLC1A5.
Collapse
Affiliation(s)
- Xiaobao Fan
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Medical College of Xi 'an Jiaotong University, Xi'an City, Shaanxi Province, 710061, China; Nephrotic Hemodialysis Center, Shaanxi Provincial People's Hospital, Xi'an City, Shaanxi Province, 710068, China
| | - Zhenjiang Li
- Nephrotic Hemodialysis Center, Shaanxi Provincial People's Hospital, Xi'an City, Shaanxi Province, 710068, China
| | - Xiaoming Wang
- Nephrotic Hemodialysis Center, Shaanxi Provincial People's Hospital, Xi'an City, Shaanxi Province, 710068, China
| | - Jing Wang
- Nephrotic Hemodialysis Center, Shaanxi Provincial People's Hospital, Xi'an City, Shaanxi Province, 710068, China
| | - Zhiming Hao
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Medical College of Xi 'an Jiaotong University, Xi'an City, Shaanxi Province, 710061, China.
| |
Collapse
|
63
|
Affiliation(s)
- Allison C. Ostriker
- Departments of Medicine and Pharmacology, Yale University School of Medicine
| | - Kathleen A. Martin
- Departments of Medicine and Pharmacology, Yale University School of Medicine
| |
Collapse
|
64
|
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
|