1
|
Liu Z, Wang H, Liang Y, Liu M, Huang Q, Wang M, Zhou J, Bu Q, Zhou H, Lu L. E2F2 Reprograms Macrophage Function By Modulating Material and Energy Metabolism in the Progression of Metabolic Dysfunction-Associated Steatohepatitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2410880. [PMID: 39465673 PMCID: PMC11672278 DOI: 10.1002/advs.202410880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/30/2024] [Indexed: 10/29/2024]
Abstract
Macrophages are essential for the development of steatosis, hepatic inflammation, and fibrosis in metabolic dysfunction-associated steatohepatitis(MASH). However, the roles of macrophage E2F2 in the progression of MASH have not been elucidated. This study reveals that the expression of macrophage E2F2 is dramatically downregulated in MASH livers from mice and humans, and that this expression is adversely correlated with the severity of the disease. Myeloid-specific E2F2 depletion aggravates intrahepatic inflammation, hepatic stellate cell activation, and hepatocyte lipid accumulation during MASH progression. Mechanistically, E2F2 can inhibit the SLC7A5 transcription directly. E2F2 deficiency upregulates the expression of SLC7A5 to mediate amino acids flux, resulting in enhanced glycolysis, impaired mitochondrial function, and increased macrophages proinflammatory response in a Leu-mTORC1-dependent manner. Moreover, bioinformatics analysis and CUT &Tag assay identify the direct binding of Nrf2 to E2F2 promoter to promote its transcription and nuclear translocation. Genetic or pharmacological activation of Nrf2 effectively activates E2F2 to attenuate the MASH progression. Finally, patients treated with CDK4/6 inhibitors demonstrate reduced E2F2 activity but increased SLC7A5 activity in PBMCs. These findings indicated macrophage E2F2 suppresses MASH progression by reprogramming amino acid metabolism via SLC7A5- Leu-mTORC1 signaling pathway. Activating E2F2 holds promise as a therapeutic strategy for MASH.
Collapse
Affiliation(s)
- Zheng Liu
- Department of General Surgerythe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029China
| | - Hao Wang
- Department of Liver SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100730China
| | - Yuan Liang
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University& Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjing210029China
- School of Biological Science & Medical EngineeringSoutheast UniversityNanjing210096China
| | - Mu Liu
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University& Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjing210029China
| | - Qiyuan Huang
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University& Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjing210029China
| | - Mingming Wang
- Department of Liver SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100730China
| | - Jinren Zhou
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University& Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjing210029China
| | - Qingfa Bu
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University& Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjing210029China
| | - Haoming Zhou
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University& Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjing210029China
| | - Ling Lu
- Affiliated Hospital of Xuzhou Medical UniversityXuzhou220005China
| |
Collapse
|
2
|
da Silva AM, Yevdokimova V, Benoit YD. Sam68 is a druggable vulnerability point in cancer stem cells. Cancer Metastasis Rev 2024; 43:441-456. [PMID: 37792222 PMCID: PMC11016129 DOI: 10.1007/s10555-023-10145-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 09/27/2023] [Indexed: 10/05/2023]
Abstract
Sam68 (Src associated in mitosis of 68 kDa) is an RNA-binding and multifunctional protein extensively characterized in numerous cellular functions, such as RNA processing, cell cycle regulation, kinase- and growth factor signaling. Recent investigations highlighted Sam68 as a primary target of a class of reverse-turn peptidomimetic drugs, initially developed as inhibitors of Wnt/β-catenin mediated transcription. Further investigations on such compounds revealed their capacity to selectively eliminate cancer stem cell (CSC) activity upon engaging Sam68. This work highlighted previously unappreciated roles for Sam68 in the maintenance of neoplastic self-renewal and tumor-initiating functions. Here, we discuss the implication of Sam68 in tumorigenesis, where central findings support its contribution to chromatin regulation processes essential to CSCs. We also review advances in CSC-targeting drug discovery aiming to modulate Sam68 cellular distribution and protein-protein interactions. Ultimately, Sam68 constitutes a vulnerability point of CSCs and an attractive therapeutic target to impede neoplastic stemness in human tumors.
Collapse
Affiliation(s)
- Amanda Mendes da Silva
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Veronika Yevdokimova
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Yannick D Benoit
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
- School of Pharmaceutical Sciences, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
3
|
Qiao A, Ma W, Jiang Y, Han C, Yan B, Zhou J, Qin G. Hepatic Sam68 Regulates Systemic Glucose Homeostasis and Insulin Sensitivity. Int J Mol Sci 2022; 23:ijms231911469. [PMID: 36232770 PMCID: PMC9569775 DOI: 10.3390/ijms231911469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/21/2022] Open
Abstract
Hepatic glucose production (HGP) is an important component of glucose homeostasis, and deregulated HGP, particularly through gluconeogenesis, contributes to hyperglycemia and pathology of type-2 diabetes (T2D). It has been shown that the gluconeogenic gene expression is governed primarily by the transcription factor cAMP-response element (CRE)-binding protein (CREB) and its coactivator, CREB-regulated transcriptional coactivator 2 (CRTC2). Recently, we have discovered that Sam68, an adaptor protein and Src kinase substrate, potently promotes hepatic gluconeogenesis by promoting CRTC2 stability; however, the detailed mechanisms remain unclear. Here we show that in response to glucagon, Sam68 increases CREB/CRTC2 transactivity by interacting with CRTC2 in the CREB/CRTC2 complex and occupying the CRE motif of promoters, leading to gluconeogenic gene expression and glucose production. In hepatocytes, glucagon promotes Sam68 nuclear import, whereas insulin elicits its nuclear export. Furthermore, ablation of Sam68 in hepatocytes protects mice from high-fat diet (HFD)-induced hyperglycemia and significantly increased hepatic and peripheral insulin sensitivities. Thus, hepatic Sam68 potentiates CREB/CRTC2-mediated glucose production, contributes to the pathogenesis of insulin resistance, and may serve as a therapeutic target for T2D.
Collapse
Affiliation(s)
- Aijun Qiao
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
- Correspondence: (A.Q.); (G.Q.); Tel.: +205-934-6690 (G.Q.); Fax: +205-934-9101 (G.Q.)
| | - Wenxia Ma
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ying Jiang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Chaoshan Han
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Baolong Yan
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Junlan Zhou
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Gangjian Qin
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Correspondence: (A.Q.); (G.Q.); Tel.: +205-934-6690 (G.Q.); Fax: +205-934-9101 (G.Q.)
| |
Collapse
|
4
|
Liu H, Chen L, Xiao W, Liu J, Long C, Zhan W, Cui C, Yang L, Chen S. Alteration of E2F2 Expression in Governing Endothelial Cell Senescence. Genes (Basel) 2022; 13:1522. [PMID: 36140689 PMCID: PMC9498592 DOI: 10.3390/genes13091522] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 01/10/2023] Open
Abstract
Endothelial cell senescence has a vital implication for vascular dysfunction, leading to age-related cardiovascular disease, especially hypertension and atherosclerosis. E2F transcription factor 2 (E2F2) plays a critical role in cell proliferation, differentiation, and DNA damage response. Up to date, no study has ever connected E2F2 to vascular endothelial cell senescence. Here, we demonstrate that E2F2 is involved in endothelial cellular senescence. We found that E2F2 expression is decreased during the replicative senescence of human umbilical vein endothelial cells (HUVECs) and the aortas of aged mice. The knockdown of E2F2 in young HUVECs induces premature senescence characterized by an increase in senescence-associated β-galactosidase (SA-β-gal) activity, a reduction in phosphorylated endothelial nitric oxide synthase (p-eNOS) and sirtuin 1 (SIRT1), and the upregulation of senescence-associated secretory phenotype (SASP) IL-6 and IL-8. The lack of E2F2 promoted cell cycle arrest, DNA damage, and cell proliferation inhibition. Conversely, E2F2 overexpression reversed the senescence phenotype and enhanced the cellular function in the senescent cells. Furthermore, E2F2 deficiency downregulated downstream target genes including CNNA2, CDK1, and FOXM1, and overexpression restored the expression of these genes. Our findings demonstrate that E2F2 plays an indispensable role in endothelial cell senescence.
Collapse
Affiliation(s)
- Hongfei Liu
- Vascular Function Laboratory, Human Aging Research Institute and School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang 330031, China
| | - Liping Chen
- Vascular Function Laboratory, Human Aging Research Institute and School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang 330031, China
| | - Wanli Xiao
- Metabolic Control and Aging, Human Aging Research Institute and School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang 330031, China
| | - Jiankun Liu
- Aging and Vascular Diseases, Human Aging Research Institute and School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang 330031, China
| | - Changkun Long
- Vascular Function Laboratory, Human Aging Research Institute and School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang 330031, China
| | - Wenxing Zhan
- Vascular Function Laboratory, Human Aging Research Institute and School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang 330031, China
| | - Cui Cui
- Department of Ophthalmology, Handan Central Hospital, Handan 056001, China
| | - Lin Yang
- Department of Nephrology, Taikang Southwestern Medical Center, Chengdu 610213, China
| | - Shenghan Chen
- Vascular Function Laboratory, Human Aging Research Institute and School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang 330031, China
| |
Collapse
|
5
|
Shen M, Gong R, Li H, Yang Z, Wang Y, Li D. Identification of key molecular markers of acute coronary syndrome using peripheral blood transcriptome sequencing analysis and mRNA-lncRNA co-expression network construction. Bioengineered 2021; 12:12087-12106. [PMID: 34753383 PMCID: PMC8809957 DOI: 10.1080/21655979.2021.2003932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Acute coronary syndrome (ACS) is a term used to describe major cardiovascular diseases, and treatment of in-stent restenosis in patients with ACS remains a major clinical challenge. Further investigation into molecular markers of ACS may aid early diagnosis, and the treatment of ACS and post-treatment recurrence. In the present study, total RNA was extracted from the peripheral blood samples of 3 patients with ACS, 3 patients with percutaneous coronary intervention (PCI)_non-restenosis, 3 patients with PCI_restenosis and 3 healthy controls. Subsequently, RNA library construction and high-throughput sequencing were performed. DESeq2 package in R was used to screen genes that were differentially expressed between the different samples. Moreover, the intersection of the differentially expressed mRNAs (DEmRNAs) and differentially expressed long noncoding RNAs (DElncRNAs) obtained. GeneCodis4.0 was used to perform function enrichment for DEmRNAs, and lncRNA-mRNA co-expression network was constructed. The GSE60993 dataset was utilized for diagnostic analysis, and the aforementioned investigations were verified using in vitro studies. Results of the present study revealed a large number of DEmRNAs and DElncRNAs in the different groups. We selected genes in the top 10 of differential expression and also involved in the co-expression of lncRNA-mRNA for diagnostic analysis in the GSE60993 dataset. The area under curve (AUC) of PDZK1IP1 (0.747), PROK2 (0.769) and LAMP3 (0.725) were all >0.7. These results indicated that the identified mRNAs and lncRNAs may act as potential clinical biomarkers, and more specifically, PDZK1IP1, PROK2 and LAMP3 may act as potential biomarkers for the diagnosis of ACS.
Collapse
Affiliation(s)
- Ming Shen
- Department of Cardiology, The First Hospital of Hebei Medical University
| | - Rui Gong
- Department of internal medicine-Endocrinology, Children's Hospital of Hebei
| | - Haibin Li
- Department of General Medicine, the Third Hospital of Hebei Medical University
| | - Zhihui Yang
- Department of General Medicine, the Third Hospital of Hebei Medical University
| | - Yunpeng Wang
- Department of General Medicine, the Third Hospital of Hebei Medical University
| | - Dandan Li
- Department of General Medicine, the Third Hospital of Hebei Medical University
| |
Collapse
|
6
|
Qiao A, Ma W, Deng J, Zhou J, Han C, Zhang E, Boriboun C, Xu S, Zhang C, Jie C, Kim JA, Habegger KM, Qiu H, Zhao TC, Zhang J, Qin G. Ablation of Sam68 in adult mice increases thermogenesis and energy expenditure. FASEB J 2021; 35:e21772. [PMID: 34252225 DOI: 10.1096/fj.202100021r] [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: 01/20/2021] [Revised: 05/27/2021] [Accepted: 06/17/2021] [Indexed: 12/18/2022]
Abstract
Genetic deletion of Src associated in mitosis of 68kDa (Sam68), a pleiotropic adaptor protein prevents high-fat diet-induced weight gain and insulin resistance. To clarify the role of Sam68 in energy metabolism in the adult stage, we generated an inducible Sam68 knockout mice. Knockout of Sam68 was induced at the age of 7-10 weeks, and then we examined the metabolic profiles of the mice. Sam68 knockout mice gained less body weight over time and at 34 or 36 weeks old, had smaller fat mass without changes in food intake and absorption efficiency. Deletion of Sam68 in mice elevated thermogenesis, increased energy expenditure, and attenuated core-temperature drop during acute cold exposure. Furthermore, we examined younger Sam68 knockout mice at 11 weeks old before their body weights deviate, and confirmed increased energy expenditure and thermogenic gene program. Thus, Sam68 is essential for the control of adipose thermogenesis and energy homeostasis in the adult.
Collapse
Affiliation(s)
- Aijun Qiao
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Wenxia Ma
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jianxin Deng
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Junlan Zhou
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Chaoshan Han
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eric Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chan Boriboun
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shiyue Xu
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chunxiang Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chunfa Jie
- Department of Biochemistry and Nutrition, Des Moines University College of Osteopathic Medicine, Des Moines, IA, USA
| | - Jeong-A Kim
- Department of Medicine-Endocrinology, Diabetes & Metabolism, Comprehensive Diabetes Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kirk M Habegger
- Department of Medicine-Endocrinology, Diabetes & Metabolism, Comprehensive Diabetes Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hongyu Qiu
- Center for Molecular and Translational Medicine, Institute of Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Ting C Zhao
- Department of Surgery, Boston University Medical School, Roger Williams Medical Center, Providence, RI, USA
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gangjian Qin
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA.,Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
7
|
Yang L, Deng J, Ma W, Qiao A, Xu S, Yu Y, Boriboun C, Kang X, Han D, Ernst P, Zhou L, Shi J, Zhang E, Li TS, Qiu H, Nakagawa S, Blackshaw S, Zhang J, Qin G. Ablation of lncRNA Miat attenuates pathological hypertrophy and heart failure. Am J Cancer Res 2021; 11:7995-8007. [PMID: 34335976 PMCID: PMC8315059 DOI: 10.7150/thno.50990] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
Rationale: The conserved long non-coding RNA (lncRNA) myocardial infarction associate transcript (Miat) was identified for its multiple single-nucleotide polymorphisms that are strongly associated with susceptibility to MI, but its role in cardiovascular biology remains elusive. Here we investigated whether Miat regulates cardiac response to pathological hypertrophic stimuli. Methods: Both an angiotensin II (Ang II) infusion model and a transverse aortic constriction (TAC) model were used in adult WT and Miat-null knockout (Miat-KO) mice to induce pathological cardiac hypertrophy. Heart structure and function were evaluated by echocardiography and histological assessments. Gene expression in the heart was evaluated by RNA sequencing (RNA-seq), quantitative real-time RT-PCR (qRT-PCR), and Western blotting. Primary WT and Miat-KO mouse cardiomyocytes were isolated and used in Ca2+ transient and contractility measurements. Results: Continuous Ang II infusion for 4 weeks induced concentric hypertrophy in WT mice, but to a lesser extent in Miat-KO mice. Surgical TAC for 6 weeks resulted in decreased systolic function and heart failure in WT mice but not in Miat-KO mice. In both models, Miat-KO mice displayed reduced heart-weight to tibia-length ratio, cardiomyocyte cross-sectional area, cardiomyocyte apoptosis, and cardiac interstitial fibrosis and a better-preserved capillary density, as compared to WT mice. In addition, Ang II treatment led to significantly reduced mRNA and protein expression of the Ca2+ cycling genes Sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a (SERCA2a) and ryanodine receptor 2 (RyR2) and a dramatic increase in global RNA splicing events in the left ventricle (LV) of WT mice, and these changes were largely blunted in Miat-KO mice. Consistently, cardiomyocytes isolated from Miat-KO mice demonstrated more efficient Ca2+ cycling and greater contractility. Conclusions: Ablation of Miat attenuates pathological hypertrophy and heart failure, in part, by enhancing cardiomyocyte contractility.
Collapse
|
8
|
Qiao A, Zhou J, Xu S, Ma W, Boriboun C, Kim T, Yan B, Deng J, Yang L, Zhang E, Song Y, Ma YC, Richard S, Zhang C, Qiu H, Habegger KM, Zhang J, Qin G. Sam68 promotes hepatic gluconeogenesis via CRTC2. Nat Commun 2021; 12:3340. [PMID: 34099657 PMCID: PMC8185084 DOI: 10.1038/s41467-021-23624-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 05/05/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatic gluconeogenesis is essential for glucose homeostasis and also a therapeutic target for type 2 diabetes, but its mechanism is incompletely understood. Here, we report that Sam68, an RNA-binding adaptor protein and Src kinase substrate, is a novel regulator of hepatic gluconeogenesis. Both global and hepatic deletions of Sam68 significantly reduce blood glucose levels and the glucagon-induced expression of gluconeogenic genes. Protein, but not mRNA, levels of CRTC2, a crucial transcriptional regulator of gluconeogenesis, are >50% lower in Sam68-deficient hepatocytes than in wild-type hepatocytes. Sam68 interacts with CRTC2 and reduces CRTC2 ubiquitination. However, truncated mutants of Sam68 that lack the C- (Sam68ΔC) or N-terminal (Sam68ΔN) domains fails to bind CRTC2 or to stabilize CRTC2 protein, respectively, and transgenic Sam68ΔN mice recapitulate the blood-glucose and gluconeogenesis profile of Sam68-deficient mice. Hepatic Sam68 expression is also upregulated in patients with diabetes and in two diabetic mouse models, while hepatocyte-specific Sam68 deficiencies alleviate diabetic hyperglycemia and improves insulin sensitivity in mice. Thus, our results identify a role for Sam68 in hepatic gluconeogenesis, and Sam68 may represent a therapeutic target for diabetes. Hepatic gluconeogenesis is important for glucose homeostasis and a therapeutic target for type 2 diabetes. Here, the authors show that the RNA-binding adaptor protein Sam68 promotes the expression level of gluconeogenic genes and increases blood glucose levels by stabilizing the transcriptional coactivator CRTC2, while hepatic Sam68 deletion alleviates hyperglycemia in mice.
Collapse
Affiliation(s)
- Aijun Qiao
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL, USA
| | - Junlan Zhou
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Shiyue Xu
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL, USA
| | - Wenxia Ma
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL, USA
| | - Chan Boriboun
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL, USA
| | - Teayoun Kim
- Department of Medicine - Endocrinology, Diabetes & Metabolism, University of Alabama at Birmingham, School of Medicine, Birmingham, AL, USA
| | - Baolong Yan
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL, USA
| | - Jianxin Deng
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL, USA
| | - Liu Yang
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL, USA
| | - Eric Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL, USA
| | - Yuhua Song
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL, USA
| | - Yongchao C Ma
- Departments of Pediatrics, Neurology and Physiology, Northwestern University Feinberg School of Medicine, Anne & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Stephane Richard
- Lady Davis Institute for Medical Research, McGill University, Montreal, QC, Canada
| | - Chunxiang Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL, USA
| | - Hongyu Qiu
- Center for Molecular and Translational Medicine, Institute of Biomedical Science Georgia State University, Atlanta, GA, USA
| | - Kirk M Habegger
- Department of Medicine - Endocrinology, Diabetes & Metabolism, University of Alabama at Birmingham, School of Medicine, Birmingham, AL, USA
| | - Jianyi Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL, USA
| | - Gangjian Qin
- Department of Biomedical Engineering, University of Alabama at Birmingham, School of Medicine and School of Engineering, Birmingham, AL, USA. .,Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
9
|
Tseng CC, Wong MC, Liao WT, Chen CJ, Lee SC, Yen JH, Chang SJ. Genetic Variants in Transcription Factor Binding Sites in Humans: Triggered by Natural Selection and Triggers of Diseases. Int J Mol Sci 2021; 22:ijms22084187. [PMID: 33919522 PMCID: PMC8073710 DOI: 10.3390/ijms22084187] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/14/2022] Open
Abstract
Variants of transcription factor binding sites (TFBSs) constitute an important part of the human genome. Current evidence demonstrates close links between nucleotides within TFBSs and gene expression. There are multiple pathways through which genomic sequences located in TFBSs regulate gene expression, and recent genome-wide association studies have shown the biological significance of TFBS variation in human phenotypes. However, numerous challenges remain in the study of TFBS polymorphisms. This article aims to cover the current state of understanding as regards the genomic features of TFBSs and TFBS variants; the mechanisms through which TFBS variants regulate gene expression; the approaches to studying the effects of nucleotide changes that create or disrupt TFBSs; the challenges faced in studies of TFBS sequence variations; the effects of natural selection on collections of TFBSs; in addition to the insights gained from the study of TFBS alleles related to gout, its associated comorbidities (increased body mass index, chronic kidney disease, diabetes, dyslipidemia, coronary artery disease, ischemic heart disease, hypertension, hyperuricemia, osteoporosis, and prostate cancer), and the treatment responses of patients.
Collapse
Affiliation(s)
- Chia-Chun Tseng
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-C.T.); (J.-H.Y.)
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
| | - Man-Chun Wong
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Wei-Ting Liao
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
- Correspondence: (W.-T.L.); (S.-J.C.); Tel.: +886-7-3121101 (W.-T.L.); +886-7-5916679 (S.-J.C.); Fax:+886-7-3125339 (W.-T.L.); +886-7-5919264 (S.-J.C.)
| | - Chung-Jen Chen
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan;
| | - Su-Chen Lee
- Laboratory Diagnosis of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Jeng-Hsien Yen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-C.T.); (J.-H.Y.)
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
- Department of Biological Science and Technology, National Chiao-Tung University, Hsinchu 30010, Taiwan
| | - Shun-Jen Chang
- Department of Kinesiology, Health and Leisure Studies, National University of Kaohsiung, Kaohsiung 81148, Taiwan
- Correspondence: (W.-T.L.); (S.-J.C.); Tel.: +886-7-3121101 (W.-T.L.); +886-7-5916679 (S.-J.C.); Fax:+886-7-3125339 (W.-T.L.); +886-7-5919264 (S.-J.C.)
| |
Collapse
|
10
|
Resveratrol Attenuates High Glucose-Induced Vascular Endothelial Cell Injury by Activating the E2F3 Pathway. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6173618. [PMID: 32420356 PMCID: PMC7204347 DOI: 10.1155/2020/6173618] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 03/21/2020] [Accepted: 04/10/2020] [Indexed: 11/22/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is the most common metabolic disease. High glucose-induced macrovascular disease and microangiopathy are major complications of diabetes. E2F3, a member of the E2F transcription factor family, is closely related to cardiovascular diseases. Resveratrol, a nonflavonoid polyphenolic compound widely found in plants, has been shown to have cardiovascular protection. However, there are few studies on whether resveratrol can effectively treat diabetic angiopathy, and the specific mechanism involved needs further study. This study investigated whether E2F3 transcription factors are involved in the process of vascular endothelial injury induced by high glucose and observed its effects on the proliferation of vascular endothelial cells. Then, it analyzed whether resveratrol can inhibit high glucose-induced vascular endothelial cell injury by regulating the E2F3 pathway. We demonstrated that the expression level of the E2F3 transcription factor was significantly inhibited in high glucose state. Resveratrol inhibited high glucose-induced vascular endothelial cell injury by upregulating the E2F3 pathway. High glucose can induce vascular endothelial injury by inhibiting E2F3 gene expression, while resveratrol can inhibit high glucose-induced vascular endothelial injury by activating the E2F3 pathway.
Collapse
|
11
|
Belloni E, Di Matteo A, Pradella D, Vacca M, Wyatt CDR, Alfieri R, Maffia A, Sabbioneda S, Ghigna C. Gene Expression Profiles Controlled by the Alternative Splicing Factor Nova2 in Endothelial Cells. Cells 2019; 8:cells8121498. [PMID: 31771184 PMCID: PMC6953062 DOI: 10.3390/cells8121498] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/11/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023] Open
Abstract
Alternative splicing (AS) plays an important role in expanding the complexity of the human genome through the production of specialized proteins regulating organ development and physiological functions, as well as contributing to several pathological conditions. How AS programs impact on the signaling pathways controlling endothelial cell (EC) functions and vascular development is largely unknown. Here we identified, through RNA-seq, changes in mRNA steady-state levels in ECs caused by the neuro-oncological ventral antigen 2 (Nova2), a key AS regulator of the vascular morphogenesis. Bioinformatics analyses identified significant enrichment for genes regulated by peroxisome proliferator-activated receptor-gamma (Ppar-γ) and E2F1 transcription factors. We also showed that Nova2 in ECs controlled the AS profiles of Ppar-γ and E2F dimerization partner 2 (Tfdp2), thus generating different protein isoforms with distinct function (Ppar-γ) or subcellular localization (Tfdp2). Collectively, our results supported a mechanism whereby Nova2 integrated splicing decisions in order to regulate Ppar-γ and E2F1 activities. Our data added a layer to the sequential series of events controlled by Nova2 in ECs to orchestrate vascular biology.
Collapse
Affiliation(s)
- Elisa Belloni
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Anna Di Matteo
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Davide Pradella
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Margherita Vacca
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Christopher D. R. Wyatt
- Centre for Biodiversity and Environment Research, University College London, Gower Street, London WC1E 6BT, UK
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr Aiguader 88, 08003 Barcelona, Spain
- Universitat Pompeu Fabra, Plaça de la Mercè, 10-12, 08002 Barcelona, Spain
| | - Roberta Alfieri
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Antonio Maffia
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Simone Sabbioneda
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
| | - Claudia Ghigna
- Istituto di Genetica Molecolare, “Luigi Luca Cavalli-Sforza”, Consiglio Nazionale delle Ricerche, via Abbiategrasso 207, 27100 Pavia, Italy; (E.B.); (A.D.M.); (D.P.); (M.V.); (R.A.); (A.M.); (S.S.)
- Correspondence:
| |
Collapse
|
12
|
Han S, Xu S, Zhou J, Qiao A, Boriboun C, Ma W, Li H, Biyashev D, Yang L, Zhang E, Liu Q, Jiang S, Zhao TC, Krishnamurthy P, Zhang C, Richard S, Qiu H, Zhang J, Qin G. Sam68 impedes the recovery of arterial injury by augmenting inflammatory response. J Mol Cell Cardiol 2019; 137:82-92. [PMID: 31639388 DOI: 10.1016/j.yjmcc.2019.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 09/23/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The role of Src-associated-in-mitosis-68-kDa (Sam68) in cardiovascular biology has not been studied. A recent report suggests that Sam68 promotes TNF-α-induced NF-κB activation in fibroblasts. Here we sought to dissect the molecular mechanism by which Sam68 regulates NF-κB signaling and its functional significance in vascular injury. APPROACH AND RESULTS The endothelial denudation injury was induced in the carotid artery of Sam68-null (Sam68-/-) and WT mice. Sam68-/- mice displayed an accelerated re-endothelialization and attenuated neointima hyperplasia, which was associated with a reduced macrophage infiltration and lowered expression of pro-inflammatory cytokines in the injured vessels. Remarkably, the ameliorated vascular remodeling was recapitulated in WT mice after receiving transplantation of bone marrow (BM) from Sam68-/- mice, suggesting the effect was attributable to BM-derived inflammatory cells. In cultured Raw264.7 macrophages, knockdown of Sam68 resulted in a significant reduction in the TNF-α-induced expression of TNF-α, IL-1β, and IL-6 and in the level of nuclear phospho-p65, indicating attenuated NF-κB activation; and these results were confirmed in peritoneal and BM-derived macrophages of Sam68-/- vs. WT mice. Furthermore, co-immunoprecipitation and mass-spectrometry identified Filamin A (FLNA) as a novel Sam68-interacting protein upon TNF-α treatment. Loss- and gain-of-function experiments suggest that Sam68 and FLNA are mutually dependent for NF-κB activation and pro-inflammatory cytokine expression, and that the N-terminus of Sam68 is required for TRAF2-FLNA interaction. CONCLUSIONS Sam68 promotes pro-inflammatory response in injured arteries and impedes recovery by interacting with FLNA to stabilize TRAF2 on the cytoskeleton and consequently potentiate NF-κB signaling.
Collapse
Affiliation(s)
- Shuling Han
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Shiyue Xu
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Junlan Zhou
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Aijun Qiao
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Chan Boriboun
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Wenxia Ma
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Huadong Li
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Dauren Biyashev
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Liu Yang
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Eric Zhang
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Qinghua Liu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, Hubei, China
| | - Shayi Jiang
- Department of Hematology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 20062, China
| | - Ting C Zhao
- Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Providence, RI 02908, USA
| | - Prasanna Krishnamurthy
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Chunxiang Zhang
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Stéphane Richard
- Lady Davis Institute for Medical Research, McGill University, Montreal, Quebec, Canada
| | - Hongyu Qiu
- Center of Molecular and Translational Medicine, Institution of Biomedical Science, Georgia State University, Atlanta, GA 30303, USA
| | - Jianyi Zhang
- Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gangjian Qin
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Molecular Cardiology Program, Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
13
|
Boosani CS, Dhar K, Agrawal DK. Down-regulation of hsa-miR-1264 contributes to DNMT1-mediated silencing of SOCS3. Mol Biol Rep 2016; 42:1365-76. [PMID: 26047583 DOI: 10.1007/s11033-015-3882-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Previously we found decreased expression of SOCS3 in neointimal hyperplastic region following balloon angioplasty in atherosclerotic micro swine. In our recent in vitro studies using human coronary artery smooth muscle cells (HCASMC), we observed the inhibition of SOCS3 expression in the presence of both TNF-α and IGF-1, correlating with the in vivo findings in microswine. We also reported that two independent mechanisms, JAK/STAT3/NFκB and promoter methylation of SOCS3 were responsible for TNF-α and IGF-1 induced SOCS3 inhibition. In this study, using miRNA array and gene expression approaches, we explored the molecular mechanisms involved in the above SOCS3 repression and identified several miRNAs that are associated with the regulation of SOCS3 expression. Our miRNA expression profiling revealed profound down-regulation of two specific miRNAs, hsa-miR-758 and hsa-miR-1264, whose expression levels were decreased by 8-10 folds in HCASMCs that were treated with both TNF-α and IGF-1. This was accompanied with a significant up-regulation of three specific miRNAs, hsa-miR-155, hsa-miR-146b-5p and hsa-miR-146a, which showed about 3-7 fold increases in their expression levels. Importantly, we also found that the miRNA hsa-miR-1264 targets DNA methyltransferase-1 (DNMT1) transcripts by binding to its 3'UTR region to affect its expression. Expression of hsa-miR-1264 in HCASMCs not only resulted in decreased DNMT1 mRNA transcripts but it also increased SOCS3 expression. The treatment with TNF-α and IGF-1 resulted in drastic decrease in hsa-miR-1264 levels with no change in the expression of DNMT1. Consequently, the DNMT1 activity caused hypermethylation in the CpG island of the SOCS3 promoter region and inhibited its expression. This could be a causative epigenetic mechanism associated with TNF-α and IGF-1 induced smooth muscle cell proliferation involved in the pathogenesis of coronary artery hyperplasia and restenosis.
Collapse
Affiliation(s)
- Chandra S Boosani
- Department of Biomedical Sciences, School of Medicine Creighton University, 2500 California Plaza, Omaha, NE, 68178, USA
| | | | | |
Collapse
|
14
|
Boosani CS, Agrawal DK. Methylation and microRNA-mediated epigenetic regulation of SOCS3. Mol Biol Rep 2015; 42:853-72. [PMID: 25682267 DOI: 10.1007/s11033-015-3860-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Epigenetic gene silencing of several genes causes different pathological conditions in humans, and DNA methylation has been identified as one of the key mechanisms that underlie this evolutionarily conserved phenomenon associated with developmental and pathological gene regulation. Recent advances in the miRNA technology with high throughput analysis of gene regulation further increased our understanding on the role of miRNAs regulating multiple gene expression. There is increasing evidence supporting that the miRNAs not only regulate gene expression but they also are involved in the hypermethylation of promoter sequences, which cumulatively contributes to the epigenetic gene silencing. Here, we critically evaluated the recent progress on the transcriptional regulation of an important suppressor protein that inhibits cytokine-mediated signaling, SOCS3, whose expression is directly regulated both by promoter methylation and also by microRNAs, affecting its vital cell regulating functions. SOCS3 was identified as a potent inhibitor of Jak/Stat signaling pathway which is frequently upregulated in several pathologies, including cardiovascular disease, cancer, diabetes, viral infections, and the expression of SOCS3 was inhibited or greatly reduced due to hypermethylation of the CpG islands in its promoter region or suppression of its expression by different microRNAs. Additionally, we discuss key intracellular signaling pathways regulated by SOCS3 involving cellular events, including cell proliferation, cell growth, cell migration and apoptosis. Identification of the pathway intermediates as specific targets would not only aid in the development of novel therapeutic drugs, but, would also assist in developing new treatment strategies that could successfully be employed in combination therapy to target multiple signaling pathways.
Collapse
Affiliation(s)
- Chandra S Boosani
- Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | | |
Collapse
|
15
|
Zhou J, Cheng M, Boriboun C, Ardehali MM, Jiang C, Liu Q, Han S, Goukassian DA, Tang YL, Zhao TC, Zhao M, Cai L, Richard S, Kishore R, Qin G. Inhibition of Sam68 triggers adipose tissue browning. J Endocrinol 2015; 225:181-9. [PMID: 25934704 PMCID: PMC4482239 DOI: 10.1530/joe-14-0727] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/22/2015] [Indexed: 12/12/2022]
Abstract
Obesity is associated with insulin resistance and type 2 diabetes; molecular mechanisms that promote energy expenditure can be utilized for effective therapy. Src-associated in mitosis of 68 kDa (Sam68) is potentially significant, because knockout (KO) of Sam68 leads to markedly reduced adiposity. In the present study, we sought to determine the mechanism by which Sam68 regulates adiposity and energy homeostasis. We first found that Sam68 KO mice have a significantly reduced body weight as compared to controls, and the difference is explained entirely by decreased adiposity. Interestingly, these effects were not mediated by a difference in food intake; rather, they were associated with enhanced physical activity. When they were fed a high-fat diet, Sam68 KO mice gained much less body weight and fat mass than their WT littermates did, and they displayed an improved glucose and insulin tolerance. In Sam68 KO mice, the brown adipose tissue (BAT), inguinal, and epididymal depots were smaller, and their adipocytes were less hypertrophied as compared to their WT littermates. The BAT of Sam68 KO mice exhibited reduced lipid stores and expressed higher levels of Ucp1 and key thermogenic and fatty acid oxidation genes. Similarly, depots of inguinal and epididymal white adipose tissue (WAT) in Sam68 KO mice appeared browner, their multilocular Ucp1-positive cells were much more abundant, and the expression of Ucp1, Cidea, Prdm16, and Ppargc1a genes was greater as compared to WT controls, which suggests that the loss of Sam68 also promotes WAT browning. Furthermore, in all of the fat depots of the Sam68 KO mice, the expression of M2 macrophage markers was up-regulated, and that of M1 markers was down-regulated. Thus, Sam68 plays a crucial role in controlling thermogenesis and may be targeted to combat obesity and associated disorders.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Adipogenesis
- Adipose Tissue, Brown/cytology
- Adipose Tissue, Brown/immunology
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/pathology
- Adipose Tissue, White/cytology
- Adipose Tissue, White/immunology
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/pathology
- Adiposity
- Animals
- Behavior, Animal
- Cell Size
- Disease Resistance
- Energy Intake
- Energy Metabolism
- Gene Expression Regulation
- Heterozygote
- Insulin Resistance
- Ion Channels/biosynthesis
- Macrophages/immunology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondrial Proteins/biosynthesis
- Motor Activity
- Obesity/immunology
- Obesity/metabolism
- Obesity/pathology
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Thermogenesis
- Uncoupling Protein 1
Collapse
Affiliation(s)
- Junlan Zhou
- Department of Medicine-Cardiology Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Tarry 14-721, Chicago, Illinois 60611, USA Department of Cardiology Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China Department of Biochemistry University of Ottawa, Ottawa, Ontario, Canada Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China GeneSys Research Institute CardioVascular Research Center, Tufts University School of Medicine, Boston, Massachusetts, USA Department of Medicine Medical College of Georgia, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA Department of Surgery Roger Williams Medical Center, Boston University Medical School, Providence, Rhode Island, USA Kosair Children Hospital Research Institute Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA Lady Davis Institute for Medical Research McGill University, Montreal, Quebec, Canada Center for Translational Medicine Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Min Cheng
- Department of Medicine-Cardiology Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Tarry 14-721, Chicago, Illinois 60611, USA Department of Cardiology Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China Department of Biochemistry University of Ottawa, Ottawa, Ontario, Canada Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China GeneSys Research Institute CardioVascular Research Center, Tufts University School of Medicine, Boston, Massachusetts, USA Department of Medicine Medical College of Georgia, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA Department of Surgery Roger Williams Medical Center, Boston University Medical School, Providence, Rhode Island, USA Kosair Children Hospital Research Institute Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA Lady Davis Institute for Medical Research McGill University, Montreal, Quebec, Canada Center for Translational Medicine Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Chan Boriboun
- Department of Medicine-Cardiology Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Tarry 14-721, Chicago, Illinois 60611, USA Department of Cardiology Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China Department of Biochemistry University of Ottawa, Ottawa, Ontario, Canada Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China GeneSys Research Institute CardioVascular Research Center, Tufts University School of Medicine, Boston, Massachusetts, USA Department of Medicine Medical College of Georgia, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA Department of Surgery Roger Williams Medical Center, Boston University Medical School, Providence, Rhode Island, USA Kosair Children Hospital Research Institute Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA Lady Davis Institute for Medical Research McGill University, Montreal, Quebec, Canada Center for Translational Medicine Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Mariam M Ardehali
- Department of Medicine-Cardiology Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Tarry 14-721, Chicago, Illinois 60611, USA Department of Cardiology Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China Department of Biochemistry University of Ottawa, Ottawa, Ontario, Canada Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China GeneSys Research Institute CardioVascular Research Center, Tufts University School of Medicine, Boston, Massachusetts, USA Department of Medicine Medical College of Georgia, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA Department of Surgery Roger Williams Medical Center, Boston University Medical School, Providence, Rhode Island, USA Kosair Children Hospital Research Institute Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA Lady Davis Institute for Medical Research McGill University, Montreal, Quebec, Canada Center for Translational Medicine Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Changfei Jiang
- Department of Medicine-Cardiology Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Tarry 14-721, Chicago, Illinois 60611, USA Department of Cardiology Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China Department of Biochemistry University of Ottawa, Ottawa, Ontario, Canada Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China GeneSys Research Institute CardioVascular Research Center, Tufts University School of Medicine, Boston, Massachusetts, USA Department of Medicine Medical College of Georgia, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA Department of Surgery Roger Williams Medical Center, Boston University Medical School, Providence, Rhode Island, USA Kosair Children Hospital Research Institute Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA Lady Davis Institute for Medical Research McGill University, Montreal, Quebec, Canada Center for Translational Medicine Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Qinghua Liu
- Department of Medicine-Cardiology Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Tarry 14-721, Chicago, Illinois 60611, USA Department of Cardiology Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China Department of Biochemistry University of Ottawa, Ottawa, Ontario, Canada Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China GeneSys Research Institute CardioVascular Research Center, Tufts University School of Medicine, Boston, Massachusetts, USA Department of Medicine Medical College of Georgia, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA Department of Surgery Roger Williams Medical Center, Boston University Medical School, Providence, Rhode Island, USA Kosair Children Hospital Research Institute Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA Lady Davis Institute for Medical Research McGill University, Montreal, Quebec, Canada Center for Translational Medicine Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Shuling Han
- Department of Medicine-Cardiology Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Tarry 14-721, Chicago, Illinois 60611, USA Department of Cardiology Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China Department of Biochemistry University of Ottawa, Ottawa, Ontario, Canada Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China GeneSys Research Institute CardioVascular Research Center, Tufts University School of Medicine, Boston, Massachusetts, USA Department of Medicine Medical College of Georgia, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA Department of Surgery Roger Williams Medical Center, Boston University Medical School, Providence, Rhode Island, USA Kosair Children Hospital Research Institute Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA Lady Davis Institute for Medical Research McGill University, Montreal, Quebec, Canada Center for Translational Medicine Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - David A Goukassian
- Department of Medicine-Cardiology Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Tarry 14-721, Chicago, Illinois 60611, USA Department of Cardiology Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China Department of Biochemistry University of Ottawa, Ottawa, Ontario, Canada Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China GeneSys Research Institute CardioVascular Research Center, Tufts University School of Medicine, Boston, Massachusetts, USA Department of Medicine Medical College of Georgia, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA Department of Surgery Roger Williams Medical Center, Boston University Medical School, Providence, Rhode Island, USA Kosair Children Hospital Research Institute Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA Lady Davis Institute for Medical Research McGill University, Montreal, Quebec, Canada Center for Translational Medicine Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Yao-Liang Tang
- Department of Medicine-Cardiology Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Tarry 14-721, Chicago, Illinois 60611, USA Department of Cardiology Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China Department of Biochemistry University of Ottawa, Ottawa, Ontario, Canada Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China GeneSys Research Institute CardioVascular Research Center, Tufts University School of Medicine, Boston, Massachusetts, USA Department of Medicine Medical College of Georgia, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA Department of Surgery Roger Williams Medical Center, Boston University Medical School, Providence, Rhode Island, USA Kosair Children Hospital Research Institute Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA Lady Davis Institute for Medical Research McGill University, Montreal, Quebec, Canada Center for Translational Medicine Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ting C Zhao
- Department of Medicine-Cardiology Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Tarry 14-721, Chicago, Illinois 60611, USA Department of Cardiology Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China Department of Biochemistry University of Ottawa, Ottawa, Ontario, Canada Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China GeneSys Research Institute CardioVascular Research Center, Tufts University School of Medicine, Boston, Massachusetts, USA Department of Medicine Medical College of Georgia, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA Department of Surgery Roger Williams Medical Center, Boston University Medical School, Providence, Rhode Island, USA Kosair Children Hospital Research Institute Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA Lady Davis Institute for Medical Research McGill University, Montreal, Quebec, Canada Center for Translational Medicine Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ming Zhao
- Department of Medicine-Cardiology Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Tarry 14-721, Chicago, Illinois 60611, USA Department of Cardiology Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China Department of Biochemistry University of Ottawa, Ottawa, Ontario, Canada Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China GeneSys Research Institute CardioVascular Research Center, Tufts University School of Medicine, Boston, Massachusetts, USA Department of Medicine Medical College of Georgia, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA Department of Surgery Roger Williams Medical Center, Boston University Medical School, Providence, Rhode Island, USA Kosair Children Hospital Research Institute Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA Lady Davis Institute for Medical Research McGill University, Montreal, Quebec, Canada Center for Translational Medicine Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Lu Cai
- Department of Medicine-Cardiology Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Tarry 14-721, Chicago, Illinois 60611, USA Department of Cardiology Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China Department of Biochemistry University of Ottawa, Ottawa, Ontario, Canada Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China GeneSys Research Institute CardioVascular Research Center, Tufts University School of Medicine, Boston, Massachusetts, USA Department of Medicine Medical College of Georgia, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA Department of Surgery Roger Williams Medical Center, Boston University Medical School, Providence, Rhode Island, USA Kosair Children Hospital Research Institute Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA Lady Davis Institute for Medical Research McGill University, Montreal, Quebec, Canada Center for Translational Medicine Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Stéphane Richard
- Department of Medicine-Cardiology Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Tarry 14-721, Chicago, Illinois 60611, USA Department of Cardiology Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China Department of Biochemistry University of Ottawa, Ottawa, Ontario, Canada Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China GeneSys Research Institute CardioVascular Research Center, Tufts University School of Medicine, Boston, Massachusetts, USA Department of Medicine Medical College of Georgia, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA Department of Surgery Roger Williams Medical Center, Boston University Medical School, Providence, Rhode Island, USA Kosair Children Hospital Research Institute Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA Lady Davis Institute for Medical Research McGill University, Montreal, Quebec, Canada Center for Translational Medicine Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Raj Kishore
- Department of Medicine-Cardiology Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Tarry 14-721, Chicago, Illinois 60611, USA Department of Cardiology Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China Department of Biochemistry University of Ottawa, Ottawa, Ontario, Canada Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China GeneSys Research Institute CardioVascular Research Center, Tufts University School of Medicine, Boston, Massachusetts, USA Department of Medicine Medical College of Georgia, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA Department of Surgery Roger Williams Medical Center, Boston University Medical School, Providence, Rhode Island, USA Kosair Children Hospital Research Institute Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA Lady Davis Institute for Medical Research McGill University, Montreal, Quebec, Canada Center for Translational Medicine Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Gangjian Qin
- Department of Medicine-Cardiology Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Tarry 14-721, Chicago, Illinois 60611, USA Department of Cardiology Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China Department of Biochemistry University of Ottawa, Ottawa, Ontario, Canada Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China GeneSys Research Institute CardioVascular Research Center, Tufts University School of Medicine, Boston, Massachusetts, USA Department of Medicine Medical College of Georgia, Vascular Biology Center, Georgia Regents University, Augusta, Georgia, USA Department of Surgery Roger Williams Medical Center, Boston University Medical School, Providence, Rhode Island, USA Kosair Children Hospital Research Institute Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA Lady Davis Institute for Medical Research McGill University, Montreal, Quebec, Canada Center for Translational Medicine Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
16
|
Qin B, Zhou J. Src Family Kinases (SFK) Mediate Angiotensin II-Induced Myosin Light Chain Phosphorylation and Hypertension. PLoS One 2015; 10:e0127891. [PMID: 26011449 PMCID: PMC4444283 DOI: 10.1371/journal.pone.0127891] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 04/20/2015] [Indexed: 11/22/2022] Open
Abstract
Angiotensin (Ang) II is the major bioactive peptide of the renin–angiotensin system (RAS); it contributes to the pathogenesis of hypertension by inducing vascular contraction and adverse remodeling, thus elevated peripheral resistance. Ang II also activates Src family kinases (SFK) in the vascular system, which has been implicated in cell proliferation and migration. However, the role of SFK in Ang II-induced hypertension is largely unknown. In this study, we found that administration of a SFK inhibitor SU6656 markedly lowered the level of systemic BP in Ang II-treated mice, which was associated with an attenuated phosphorylation of the smooth-muscle myosin-light-chain (MLC) in the mesenteric resistant arteries. In the cultured human coronary artery smooth muscle cells (SMCs), pretreatment with SU6656 blocked Ang II-induced MLC phosphorylation and contraction. These results for the first time demonstrate that SFK directly regulate vascular contractile machinery to influence BP. Thus our study provides an additional mechanistic link between Ang II and vasoconstriction via SFK-enhanced MLC phosphorylation in SMCs, and suggests that targeted inhibition of Src may provide a new therapeutic opportunity in the treatment of hypertension.
Collapse
Affiliation(s)
- Bo Qin
- Weinberg College of Arts and Sciences, Northwestern University, Chicago, IL, United States of America
| | - Junlan Zhou
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States of America
- * E-mail:
| |
Collapse
|
17
|
Wu M, Zhou J, Cheng M, Boriboun C, Biyashev D, Wang H, Mackie A, Thorne T, Chou J, Wu Y, Chen Z, Liu Q, Yan H, Yang Y, Jie C, Tang YL, Zhao TC, Taylor RN, Kishore R, Losordo DW, Qin G. E2F1 suppresses cardiac neovascularization by down-regulating VEGF and PlGF expression. Cardiovasc Res 2014; 104:412-22. [PMID: 25341896 DOI: 10.1093/cvr/cvu222] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
AIMS The E2F transcription factors are best characterized for their roles in cell-cycle regulation, cell growth, and cell death. Here we investigated the potential role of E2F1 in cardiac neovascularization. METHODS AND RESULTS We induced myocardial infarction (MI) by ligating the left anterior descending artery in wild-type (WT) and E2F1(-/-) mice. E2F1(-/-) mice demonstrated a significantly better cardiac function and smaller infarct sizes than WT mice. At infarct border zone, capillary density and endothelial cell (EC) proliferation were greater, apoptotic ECs were fewer, levels of VEGF and placental growth factor (PlGF) were higher, and p53 level was lower in E2F1(-/-) than in WT mice. Blockade of VEGF receptor 2 (VEGFR2) signalling with the selective inhibitor SU5416 or with the VEGFR2-blocking antibody DC101 abolished the differences between E2F1(-/-) mice and WT mice in cardiac function, infarct size, capillary density, EC proliferation, and EC apoptosis. In vitro, hypoxia-induced VEGF and PlGF up-regulation was significantly greater in E2F1(-/-) than in WT cardiac fibroblasts, and E2F1 overexpression suppressed PlGF up-regulation in both WT and p53(-/-) cells; however, VEGF up-regulation was suppressed only in WT cells. E2F1 interacted with and stabilized p53 under hypoxic conditions, and both E2F1 : p53 binding and the E2F1-induced suppression of VEGF promoter activity were absent in cells that expressed an N-terminally truncated E2F1 mutant. CONCLUSION E2F1 limits cardiac neovascularization and functional recovery after MI by suppressing VEGF and PlGF up-regulation through p53-dependent and -independent mechanisms, respectively.
Collapse
Affiliation(s)
- Min Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| | - Junlan Zhou
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| | - Min Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chan Boriboun
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| | - Dauren Biyashev
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| | - Hong Wang
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| | - Alexander Mackie
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| | - Tina Thorne
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| | - Jonathan Chou
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| | - Yiping Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhishui Chen
- Organ Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qinghua Liu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan, Hubei, China
| | - Hongbin Yan
- Cardiology Department, Cardiovascular Institute and Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ya Yang
- Department of Echocardiography, Beijing Anzhen Hospital, Capital Medical University and Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Chunfa Jie
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yao-Liang Tang
- Department of Medicine, Vascular Biology Center, Medical College of Georgia/Georgia Regents University, Augusta, GA, USA
| | - Ting C Zhao
- Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, RI, USA
| | - Robert N Taylor
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Raj Kishore
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Douglas W Losordo
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| | - Gangjian Qin
- Department of Medicine-Cardiology, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Ave., Tarry 14-721, Chicago, IL 60611, USA
| |
Collapse
|
18
|
Contrasting roles of E2F2 and E2F3 in cardiac neovascularization. PLoS One 2013; 8:e65755. [PMID: 23799044 PMCID: PMC3683051 DOI: 10.1371/journal.pone.0065755] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 04/29/2013] [Indexed: 01/23/2023] Open
Abstract
Insufficient neovascularization, characterized by poor endothelial cell (EC) growth, contributes to the pathogenesis of ischemic heart disease and limits cardiac tissue preservation and regeneration. The E2F family of transcription factors are critical regulators of the genes responsible for cell-cycle progression and growth; however, the specific roles of individual E2Fs in ECs are not well understood. Here we investigated the roles of E2F2 and E2F3 in EC growth, angiogenesis, and their functional impact on myocardial infarction (MI). An endothelial-specific E2F3-deficient mouse strain VE-Cre; E2F3(fl/fl) was generated, and MI was surgically induced in VE-Cre; E2F3(fl/fl) and E2F2-null (E2F2 KO) mice and their wild-type (WT) littermates, VE-Cre; E2F3(+/+) and E2F2 WT, respectively. The cardiac function, infarct size, and vascular density were significantly better in E2F2 KO mice and significantly worse in VE-Cre; E2F3(fl/fl) mice than in their WT littermates. The loss of E2F2 expression was associated with an increase in the proliferation of ECs both in vivo and in vitro, while the loss of E2F3 expression led to declines in EC proliferation. Thus, E2F3 promotes while E2F2 suppresses ischemic cardiac repair through corresponding changes in EC proliferation; and differential targeting of specific E2F members may provide a novel strategy for therapeutic angiogenesis of ischemic heart disease.
Collapse
|
19
|
Zhou J, Cheng M, Liao YH, Hu Y, Wu M, Wang Q, Qin B, Wang H, Zhu Y, Gao XM, Goukassian D, Zhao TC, Tang YL, Kishore R, Qin G. Rosuvastatin enhances angiogenesis via eNOS-dependent mobilization of endothelial progenitor cells. PLoS One 2013; 8:e63126. [PMID: 23704894 PMCID: PMC3660394 DOI: 10.1371/journal.pone.0063126] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 03/29/2013] [Indexed: 01/08/2023] Open
Abstract
Circulating endothelial progenitor cells (circEPCs) of bone marrow (BM) origin contribute to postnatal neovascularization and represent a potential therapeutic target for ischemic disease. Statins are beneficial for ischemia disease and have been implicated to increase neovascularization via mechanisms independent of lipid lowering. However, the effect of Statins on EPC function is not completely understood. Here we sought to investigate the effects of Rosuvastatin (Ros) on EPC mobilization and EPC-mediated neovascularization during ischemic injury. In a mouse model of surgically-induced hindlimb ischemia (HLI), treatment of mice with low dose (0.1 mg/kg) but not high dose (5 mg/kg) significantly increased capillary density and accelerated blood flow recovery, as compared to saline-treated group. When HLI was induced in mice that had received Tie2/LacZ BM transplantation, Ros treatment led a significantly larger amount of endothelial cells (ECs) of BM origin incorporated at ischemic sites than saline. After treatment of mice with a single low dose of Ros, circEPCs significantly increased from 2 h, peaked at 4 h, declined until 8 h. In a growth-factor reduced Matrigel plug-in assay, Ros treatment for 5 d induced endothelial lineage differentiation in vivo. Interestingly, the enhanced circEPCs and post-HLI neovascularization stimulated by Ros were blunted in mice deficient in endothelial nitric oxide synthase (eNOS), and Ros increased p-Akt/p-eNOS levels in EPCs in vitro, indicating these effects of Ros are dependent on eNOS activity. We conclude that Ros increases circEPCs and promotes their de novo differentiation through eNOS pathway.
Collapse
Affiliation(s)
- Junlan Zhou
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Min Cheng
- Department of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Yu-Hua Liao
- Department of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Yu Hu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Min Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Qing Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Bo Qin
- Weinberg College of Arts and Sciences, Northwestern, Chicago, Illinois, United States of America
| | - Hong Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Yan Zhu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - Xiu-Mei Gao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, P. R. China
| | - David Goukassian
- CardioVascular Systems Biology, Steward St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Ting C. Zhao
- Department of Surgery, Boston University Medical School, Roger William Medical Center, Providence, Rhode Island, United States of America
| | - Yao-Liang Tang
- Division of Cardiovascular Disease, Cardiovascular Research Center, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Raj Kishore
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Gangjian Qin
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
20
|
Zhou J, Cheng M, Wu M, Boriboun C, Jujo K, Xu S, Zhao TC, Tang YL, Kishore R, Qin G. Contrasting roles of E2F2 and E2F3 in endothelial cell growth and ischemic angiogenesis. J Mol Cell Cardiol 2013; 60:68-71. [PMID: 23603666 DOI: 10.1016/j.yjmcc.2013.04.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 03/28/2013] [Accepted: 04/10/2013] [Indexed: 01/03/2023]
Abstract
The growth of new blood vessels after ischemic injury requires endothelial cells (ECs) to divide and proliferate, and the E2F transcription factors are key regulators of the genes responsible for cell-cycle progression; however, the specific roles of individual E2Fs in ECs are largely unknown. To determine the roles of E2F2 and E2F3 in EC proliferation and the angiogenic response to ischemic injury, hind-limb ischemia was surgically induced in E2F2(-/-) mice, endothelial-specific E2F3-knockout (EndoE2F3(∆/∆)) mice, and their littermates with wild-type E2F2 and E2F3 expression. Two weeks later, Laser-Doppler perfusion measurements, capillary density, and endothelial proliferation were significantly greater in E2F2(-/-) mice and significantly lower in EndoE2F3(∆/∆) mice than in their littermates, and EndoE2F3(∆/∆) mice also developed toe and limb necrosis. The loss of E2F2 expression was associated with increases in the proliferation and G1/S-phase gene expression of isolated ECs, while the loss of E2F3 expression led to declines in these parameters. Thus E2F2 impairs, and endothelial E2F3 promotes, the angiogenic response to peripheral ischemic injury through corresponding changes in EC cell-cycle progression.
Collapse
Affiliation(s)
- Junlan Zhou
- Feinberg Cardiovascular Research Institute, Department of Medicine-Cardiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Affiliation(s)
- Joseph A. Vita
- From the Evans Department of Medicine and the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA
| |
Collapse
|
22
|
Biyashev D, Qin G. E2F and microRNA regulation of angiogenesis. AMERICAN JOURNAL OF CARDIOVASCULAR DISEASE 2011; 1:110-118. [PMID: 22200034 PMCID: PMC3244022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Accepted: 06/22/2011] [Indexed: 05/31/2023]
Abstract
E2F family of transcription factors are best known for regulating genes involved in cell cycle control, cell proliferation, tumorigenesis, and apoptosis. Recent evidences have revealed their critical involvement in modulating cellular response to hypoxia and ischemia in a variety of physiological and pathological processes. Of particular interest are findings that E2Fs act as both regulators and targets of microRNAs that govern hypoxic/ischemic angiogenesis. This review focuses on the crosstalk between E2Fs and microRNAs that have been shown to participate in the regulation of angiogenesis, hypoxia response and ischemic disease.
Collapse
Affiliation(s)
- Dauren Biyashev
- Feinberg Cardiovascular Research Institute, Department of Medicine, Cardiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | |
Collapse
|
23
|
Cheng M, Zhou J, Wu M, Boriboun C, Thorne T, Liu T, Xiang Z, Zeng Q, Tanaka T, Tang YL, Kishore R, Tomasson MH, Miller RJ, Losordo DW, Qin G. CXCR4-mediated bone marrow progenitor cell maintenance and mobilization are modulated by c-kit activity. Circ Res 2010; 107:1083-93. [PMID: 20847314 DOI: 10.1161/circresaha.110.220970] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE The mobilization of bone marrow (BM) progenitor cells (PCs) is largely governed by interactions between stromal cell-derived factor (SDF)-1 and CXC chemokine receptor (CXCR)4. Ischemic injury disrupts the SDF-1-CXCR4 interaction and releases BM PCs into the peripheral circulation, where the mobilized cells are recruited to the injured tissue and contribute to vessel growth. BM PCs can also be mobilized by the pharmacological CXCR4 antagonist AMD3100, but the other components of the SDF-1-CXCR4 signaling pathway are largely unknown. c-kit, a membrane-bound tyrosine kinase and the receptor for stem cell factor, has also been shown to play a critical role in BM PC mobilization and ischemic tissue repair. OBJECTIVE To investigate the functional interaction between SDF-1-CXCR4 signaling and c-kit activity in BM PC mobilization. METHODS AND RESULTS AMD3100 administration failed to mobilize BM PCs in mice defective in c-kit kinase activity or in mice transplanted with BM cells that expressed a constitutively active c-kit mutant. Furthermore, BM levels of phosphorylated (phospho)-c-kit declined after AMD3100 administration and after CXCR4 deletion. In cells adhering to culture plates coated with vascular cell adhesion molecule 1, SDF-1 and stem cell factor increased phospho-c-kit levels, and AMD3100 treatment suppressed SDF-1-induced, but not SCF-induced, c-kit phosphorylation. SDF-1-induced c-kit phosphorylation also required the activation of Src nonreceptor tyrosine kinase: pretreatment of cells with a selective Src inhibitor blocked both c-kit phosphorylation and the interaction between c-kit and phospho-Src. CONCLUSIONS These findings indicate that the regulation of BM PC trafficking by SDF-1 and CXCR4 is dependent on Src-mediated c-kit phosphorylation.
Collapse
Affiliation(s)
- Min Cheng
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, 303 E Chicago Avenue, Chicago, IL 60611, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|