1
|
Wu M, Zhao Y, Yang J, Yang F, Dai Y, Wang Q, Chen C, Chu X. The role of ankyrin repeat-containing proteins in epigenetic and transcriptional regulation. Cell Death Discov 2025; 11:232. [PMID: 40350474 PMCID: PMC12066720 DOI: 10.1038/s41420-025-02519-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025] Open
Abstract
Ankyrin repeat (AR) motif is one of the most abundant repeat motifs found in eukaryotic proteins. It functions in mediating protein-protein interactions and regulating numerous biological functions. Interestingly, some AR-containing proteins are involved in epigenetic and transcriptional events. Our review aims to characterize the structure and post-translational modification of AR, summarize the prominent role of AR-containing proteins in epigenetic and transcriptional events, emphasizing the crucial functions mediated by AR motifs.
Collapse
Affiliation(s)
- Meijuan Wu
- Department of Medical Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yulu Zhao
- Department of Medical Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jiahe Yang
- Department of Medical Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Fangyuan Yang
- Department of Medical Oncology, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Yeyang Dai
- Department of Medical Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qian Wang
- Department of Medical Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China
| | - Cheng Chen
- Department of Medical Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Xiaoyuan Chu
- Department of Medical Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Medical Oncology, Jinling Hospital, Nanjing Medical University, Nanjing, China.
- Department of Medical Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China.
| |
Collapse
|
2
|
Cheng L, Zhou Z, Li Q, Li W, Li X, Li G, Fan J, Yu L, Yin G. Dendronized chitosan hydrogel with GIT1 to accelerate bone defect repair through increasing local neovascular amount. Bone Rep 2023; 19:101712. [PMID: 37744736 PMCID: PMC10511783 DOI: 10.1016/j.bonr.2023.101712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/15/2023] [Accepted: 08/29/2023] [Indexed: 09/26/2023] Open
Abstract
Bone defects have long been a major healthcare issue because of the difficulties in regenerating bone mass volume and the high cost of treatment. G protein-coupled receptor kinase 2 interacting protein 1 (GIT1) has been proven to play an important role both in vascular development and in bone fracture healing. In this study, a type of thermoresponsive injectable hydrogel from oligoethylene glycol-based dendronized chitosan (G1-CS) was loaded with GIT1-plasmids (G1-CS/GIT1), and used to fill unicortical bone defects. RT-PCR analysis confirmed that G1-CS/GIT1 enhanced DNA transfection in MSCs both in vitro and in vivo. From the results of micro-CT, RT-PCR and histological analysis, it can be concluded that G1-CS/GIT1 accelerated the bone healing rate and increased the amount of neovascularization around the bone defects. In addition, an adeno-associated virus (AAV)-GIT1 was constructed to transfect mesenchymal stem cells. The results of capillary tube formation assay, immunofluorescence staining and western blot analysis proved that high expression of GIT1 induces mesenchymal stem cells to differentiate into endothelial cells. RT-PCR analysis and capillary tube formation assay confirmed that the Notch signaling pathway was activated in the differentiation process. Overall, we developed an efficient strategy through combination of injectable hydrogel and G1T1 for bone tissue engineering.
Collapse
Affiliation(s)
- Lin Cheng
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Huaihai West Road 99, Xuzhou, Jiangsu Province 221000, China
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, Jiangsu Province 210000, China
| | - Zhimin Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, Jiangsu Province 210000, China
| | - Qingqing Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, Jiangsu Province 210000, China
| | - Wen Li
- School of Materials Science and Engineering, Shanghai University, Nanchen Street 333, Shanghai 200444, China
| | - Xin Li
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Huaihai West Road 99, Xuzhou, Jiangsu Province 221000, China
| | - Gen Li
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Huaihai West Road 99, Xuzhou, Jiangsu Province 221000, China
| | - Jin Fan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, Jiangsu Province 210000, China
| | - Lipeng Yu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, Jiangsu Province 210000, China
| | - Guoyong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, Jiangsu Province 210000, China
| |
Collapse
|
3
|
Wang S, Singh M, Yang H, Morrell CN, Mohamad LA, Xu JJ, Nguyen T, Ture S, Tyrell A, Maggirwar SB, Schifitto G, Pang J. Monocyte-derived Dll4 is a novel contributor to persistent systemic inflammation in HIV patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.18.537330. [PMID: 37131726 PMCID: PMC10153122 DOI: 10.1101/2023.04.18.537330] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Background In people living with HIV (PLWH) on combination antiretroviral therapy (cART), persistent systemic inflammation is a driving force for the progression of comorbidities, such as cardiovascular and cerebrovascular diseases. In this context, monocyte- and macrophage-related inflammation rather than T cell activation is a major cause of chronic inflammation. However, the underlying mechanism of how monocytes cause persistent systemic inflammation in PLWH is elusive. Methods and Results In vitro, we demonstrated that lipopolysaccharides (LPS) or tumor necrosis factor alpha (TNFα), induced a robust increase of Delta-like ligand 4 (Dll4) mRNA and protein expression in human monocytes and Dll4 secretion (extracellular Dll4, exDll4) from monocytes. Enhanced membrane-bound Dll4 (mDll4) expression in monocytes triggered Notch1 activation to promote pro-inflammatory factors expression. Dll4 silencing and inhibition of Nocth1 activation diminished the LPS or TNFα -induced inflammation. exDll4 releases in response to cytokines occurred in monocytes but not endothelial cells or T cells. In clinical specimens, we found that PLWH, both male and female, on cART, showed a significant increase in mDll4 expression, activation of Dll4-Notch1 signaling, and inflammatory markers in monocytes. Although there was no sex effect on mDII4 in PLWH, plasma exDll4 was significantly elevated in males but not females compared to HIV uninfected individuals. Furthermore, exDll4 plasma levels paralleled with monocytes mDll4 in male PLWH. Circulating exDll4 was also positively associated with pro-inflammatory monocytes phenotype and negatively associated with classic monocytes phenotype in male PLWH. Conclusion Pro-inflammatory stimuli increase Dll4 expression and Dll4-Notch1 signaling activation in monocytes and enhance monocyte proinflammatory phenotype, contributing to persistent systemic inflammation in male and female PLWH. Therefore, monocyte mDll4 could be a potential biomarker and therapeutic target of systemic inflammation. Plasma exDll4 may also play an additional role in systemic inflammation but primarily in men.
Collapse
|
4
|
Yadunandanan Nair N, Samuel V, Ramesh L, Marib A, David DT, Sundararaman A. Actin cytoskeleton in angiogenesis. Biol Open 2022; 11:bio058899. [PMID: 36444960 PMCID: PMC9729668 DOI: 10.1242/bio.058899] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024] Open
Abstract
Actin, one of the most abundant intracellular proteins in mammalian cells, is a critical regulator of cell shape and polarity, migration, cell division, and transcriptional response. Angiogenesis, or the formation of new blood vessels in the body is a well-coordinated multi-step process. Endothelial cells lining the blood vessels acquire several new properties such as front-rear polarity, invasiveness, rapid proliferation and motility during angiogenesis. This is achieved by changes in the regulation of the actin cytoskeleton. Actin remodelling underlies the switch between the quiescent and angiogenic state of the endothelium. Actin forms endothelium-specific structures that support uniquely endothelial functions. Actin regulators at endothelial cell-cell junctions maintain the integrity of the blood-tissue barrier while permitting trans-endothelial leukocyte migration. This review focuses on endothelial actin structures and less-recognised actin-mediated endothelial functions. Readers are referred to other recent reviews for the well-recognised roles of actin in endothelial motility, barrier functions and leukocyte transmigration. Actin generates forces that are transmitted to the extracellular matrix resulting in vascular matrix remodelling. In this review, we attempt to synthesize our current understanding of the roles of actin in vascular morphogenesis. We speculate on the vascular bed specific differences in endothelial actin regulation and its role in the vast heterogeneity in endothelial morphology and function across the various tissues of our body.
Collapse
Affiliation(s)
- Nidhi Yadunandanan Nair
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Victor Samuel
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Lariza Ramesh
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Areeba Marib
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Deena T. David
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Ananthalakshmy Sundararaman
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| |
Collapse
|
5
|
Fan L, Liu H, Zhu G, Singh S, Yu Z, Wang S, Luo H, Liu S, Xu Y, Ge J, Jiang D, Pang J. Caspase-4/11 is critical for angiogenesis by repressing Notch1 signaling via inhibiting γ-secretase activity. Br J Pharmacol 2022; 179:4809-4828. [PMID: 35737588 DOI: 10.1111/bph.15904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/23/2022] [Accepted: 05/29/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Notch1 activation mediated by γ-secretase is critical for angiogenesis. GeneCards database predicted that Caspase-4 (CASP4, with murine ortholog CASP11) interacts with presenilin-1, the catalytic core of γ-secretase. Therefore, we investigated the role of CASP4/11 in angiogenesis. EXPERIMENTAL APPROACH In vivo, we studied the role of Casp11 in several angiogenesis mouse models using Casp11 wild-type and knockout mice. In vitro, we detected the effects of CASP4 on endothelial functions and Notch signaling by depleting or overexpressing CASP4 in human umbilical vein endothelial cells (HUVECs). The functional domain responsible for the binding of CASP4 and presenilin-1 was detected by mutagenesis and co-immunoprecipitation. KEY RESULTS Casp11 deficiency remarkably impaired adult angiogenesis in ischemic hindlimbs, melanoma xenografts and Matrigel plugs, but not the developmental angiogenesis of retina. Bone marrow transplantation revealed that the pro-angiogenic effect depended on CASP11 derived from non-hematopoietic cells. CASP4 expression was induced by inflammatory factors and CASP4 knockdown decreased cell viability, proliferation, migration and tube formation in HUVECs. Mechanistically, CASP4/11 deficiency increased Notch1 activation in vivo and in vitro, while CASP4 overexpression repressed Notch1 signaling in HUVECs. Moreover, CASP4 knockdown increased γ-secretase activity. γ-Secretase inhibitor DAPT restored the effects of CASP4 siRNA on Notch1 activation and angiogenesis in HUVECs. Notably, the catalytic activity of CASP4/11 was dispensable. Instead, CASP4 directly interacted with presenilin-1 through the caspase recruitment domain (CARD). CONCLUSIONS AND IMPLICATIONS These findings reveal a critical role of CASP4/11 in adult angiogenesis and make this molecule a promising therapeutic target for angiogenesis-related diseases in the future.
Collapse
Affiliation(s)
- Linlin Fan
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Hao Liu
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guofu Zhu
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shekhar Singh
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ze Yu
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shumin Wang
- Aab Cardiovascular Research Institute, Department of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Hong Luo
- Department of Medical Laboratory, College of Laboratory Medicine, Dalian Medical University, Dalian, China
| | - Shiying Liu
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yawei Xu
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Junbo Ge
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Dongyang Jiang
- Department of Cardiology, Pan-vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinjiang Pang
- Aab Cardiovascular Research Institute, Department of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| |
Collapse
|
6
|
Zhu G, Lin Y, Ge T, Singh S, Liu H, Fan L, Wang S, Rhen J, Jiang D, Lyu Y, Yin Y, Li X, Benoit DSW, Li W, Xu Y, Pang J. A novel peptide inhibitor of Dll4-Notch1 signalling and its pro-angiogenic functions. Br J Pharmacol 2022; 179:1716-1731. [PMID: 34796471 PMCID: PMC9040338 DOI: 10.1111/bph.15743] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND PURPOSE The Dll4-Notch1 signalling pathway plays an important role in sprouting angiogenesis, vascular remodelling and arterial or venous specificity. Genetic or pharmacological inhibition of Dll4-Notch1 signalling leads to excessive sprouting angiogenesis. However, transcriptional inhibitors of Dll4-Notch1 signalling have not been described. EXPERIMENTAL APPROACH We designed a new peptide targeting Notch signalling, referred to as TAT-ANK, and assessed its effects on angiogenesis. In vitro, tube formation and fibrin gel bead assay were carried out, using human umbilical vein endothelial cells (HUVECs). In vivo, Matrigel plug angiogenesis assay, a developmental retinal model and tumour models in mice were used. The mechanisms underlying TAT-ANK activity were investigated by immunochemistry, western blotting, immunoprecipitation, RT-qPCR and luciferase reporter assays. KEY RESULTS The amino acid residues 179-191 in the G-protein-coupled receptor-kinase-interacting protein-1 (GIT1-ankyrin domain) are crucial for GIT1 binding to the Notch transcription repressor, RBP-J. We designed the peptide TAT-ANK, based on residues 179-191 in GIT1. TAT-ANK significantly inhibited Dll4 expression and Notch 1 activation in HUVECs by competing with activated Notch1 to bind to RBP-J. The analyses of biological functions showed that TAT-ANK promoted angiogenesis in vitro and in vivo by inhibiting Dll4-Notch1 signalling. CONCLUSIONS AND IMPLICATIONS We synthesized and investigated the biological actions of TAT-ANK peptide, a new inhibitor of Notch signalling. This peptide will be of significant interest to research on Dll4-Notch1 signalling and to clinicians carrying out clinical trials using Notch signalling inhibitors. Furthermore, our findings will have important conceptual and therapeutic implications for angiogenesis-related diseases.
Collapse
Affiliation(s)
- Guofu Zhu
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ying Lin
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tandi Ge
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shekhar Singh
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hao Liu
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Linlin Fan
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shumin Wang
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Jordan Rhen
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Dongyang Jiang
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuyan Lyu
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yiheng Yin
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiankai Li
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Danielle S. W. Benoit
- Departments of Biomedical Engineering and Chemical Engineering, Materials Science Program, and Centers for Musculoskeletal Research and Oral Biology, University of Rochester, Rochester, New York, USA
| | - Weiming Li
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yawei Xu
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinjiang Pang
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
7
|
GIT1 protects against breast cancer growth through negative regulation of Notch. Nat Commun 2022; 13:1537. [PMID: 35318302 PMCID: PMC8940956 DOI: 10.1038/s41467-022-28631-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 01/18/2022] [Indexed: 12/20/2022] Open
Abstract
Hyperactive Notch signalling is frequently observed in breast cancer and correlates with poor prognosis. However, relatively few mutations in the core Notch signalling pathway have been identified in breast cancer, suggesting that as yet unknown mechanisms increase Notch activity. Here we show that increased expression levels of GIT1 correlate with high relapse-free survival in oestrogen receptor-negative (ER(-)) breast cancer patients and that GIT1 mediates negative regulation of Notch. GIT1 knockdown in ER(-) breast tumour cells increased signalling downstream of Notch and activity of aldehyde dehydrogenase, a predictor of poor clinical outcome. GIT1 interacts with the Notch intracellular domain (ICD) and influences signalling by inhibiting the cytoplasm-to-nucleus transport of the Notch ICD. In xenograft experiments, overexpression of GIT1 in ER(-) cells prevented or reduced Notch-driven tumour formation. These results identify GIT1 as a modulator of Notch signalling and a guardian against breast cancer growth. Notch signalling is reported to be hyperactivated in oestrogen receptor-negative (ER-) breast cancer. Here the authors show that G protein-coupled receptor kinase-interacting protein 1 (GIT1) negatively regulates Notch signalling and tumour growth in ER- breast cancer by blocking Notch ICD nuclear translocation.
Collapse
|
8
|
Wang Z, Chen J, Babicheva A, Jain PP, Rodriguez M, Ayon RJ, Ravellette KS, Wu L, Balistrieri F, Tang H, Wu X, Zhao T, Black SM, Desai AA, Garcia JGN, Sun X, Shyy JYJ, Valdez-Jasso D, Thistlethwaite PA, Makino A, Wang J, Yuan JXJ. Endothelial upregulation of mechanosensitive channel Piezo1 in pulmonary hypertension. Am J Physiol Cell Physiol 2021; 321:C1010-C1027. [PMID: 34669509 PMCID: PMC8714987 DOI: 10.1152/ajpcell.00147.2021] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/22/2021] [Accepted: 10/12/2021] [Indexed: 12/16/2022]
Abstract
Piezo is a mechanosensitive cation channel responsible for stretch-mediated Ca2+ and Na+ influx in multiple types of cells. Little is known about the functional role of Piezo1 in the lung vasculature and its potential pathogenic role in pulmonary arterial hypertension (PAH). Pulmonary arterial endothelial cells (PAECs) are constantly under mechanic stretch and shear stress that are sufficient to activate Piezo channels. Here, we report that Piezo1 is significantly upregulated in PAECs from patients with idiopathic PAH and animals with experimental pulmonary hypertension (PH) compared with normal controls. Membrane stretch by decreasing extracellular osmotic pressure or by cyclic stretch (18% CS) increases Ca2+-dependent phosphorylation (p) of AKT and ERK, and subsequently upregulates expression of Notch ligands, Jagged1/2 (Jag-1 and Jag-2), and Delta like-4 (DLL4) in PAECs. siRNA-mediated downregulation of Piezo1 significantly inhibited the stretch-mediated pAKT increase and Jag-1 upregulation, whereas downregulation of AKT by siRNA markedly attenuated the stretch-mediated Jag-1 upregulation in human PAECs. Furthermore, the mRNA and protein expression level of Piezo1 in the isolated pulmonary artery, which mainly contains pulmonary arterial smooth muscle cells (PASMCs), from animals with severe PH was also significantly higher than that from control animals. Intraperitoneal injection of a Piezo1 channel blocker, GsMTx4, ameliorated experimental PH in mice. Taken together, our study suggests that membrane stretch-mediated Ca2+ influx through Piezo1 is an important trigger for pAKT-mediated upregulation of Jag-1 in PAECs. Upregulation of the mechanosensitive channel Piezo1 and the resultant increase in the Notch ligands (Jag-1/2 and DLL4) in PAECs may play a critical pathogenic role in the development of pulmonary vascular remodeling in PAH and PH.
Collapse
Affiliation(s)
- Ziyi Wang
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiyuan Chen
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Aleksandra Babicheva
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Pritesh P Jain
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Marisela Rodriguez
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Ramon J Ayon
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Keeley S Ravellette
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Linda Wu
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Francesca Balistrieri
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Haiyang Tang
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaomin Wu
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Tengteng Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Stephen M Black
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Ankit A Desai
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
- Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Joe G N Garcia
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Xin Sun
- Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - John Y-J Shyy
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Daniela Valdez-Jasso
- Department of Bioengineering, University of California, San Diego, La Jolla, California
| | | | - Ayako Makino
- Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Jian Wang
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| |
Collapse
|
9
|
Katakia YT, Thakkar NP, Thakar S, Sakhuja A, Goyal R, Sharma H, Dave R, Mandloi A, Basu S, Nigam I, Kuncharam BVR, Chowdhury S, Majumder S. Dynamic alterations of H3K4me3 and H3K27me3 at ADAM17 and Jagged-1 gene promoters cause an inflammatory switch of endothelial cells. J Cell Physiol 2021; 237:992-1012. [PMID: 34520565 DOI: 10.1002/jcp.30579] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/25/2021] [Accepted: 08/28/2021] [Indexed: 01/01/2023]
Abstract
Histone protein modifications control the inflammatory state of many immune cells. However, how dynamic alteration in histone methylation causes endothelial inflammation and apoptosis is not clearly understood. To examine this, we explored two contrasting histone methylations; an activating histone H3 lysine 4 trimethylation (H3K4me3) and a repressive histone H3 lysine 27 trimethylation (H3K27me3) in endothelial cells (EC) undergoing inflammation. Through computer-aided reconstruction and 3D printing of the human coronary artery, we developed a unique model where EC were exposed to a pattern of oscillatory/disturbed flow as similar to in vivo conditions. Upon induction of endothelial inflammation, we detected a significant rise in H3K4me3 caused by an increase in the expression of SET1/COMPASS family of H3K4 methyltransferases, including MLL1, MLL2, and SET1B. In contrast, EC undergoing inflammation exhibited truncated H3K27me3 level engendered by EZH2 cytosolic translocation through threonine 367 phosphorylation and an increase in the expression of histone demethylating enzyme JMJD3 and UTX. Additionally, many SET1/COMPASS family of proteins, including MLL1 (C), MLL2, and WDR5, were associated with either UTX or JMJD3 or both and such association was elevated in EC upon exposure to inflammatory stimuli. Dynamic enrichment of H3K4me3 and loss of H3K27me3 at Notch-associated gene promoters caused ADAM17 and Jagged-1 derepression and abrupt Notch activation. Conversely, either reducing H3K4me3 or increasing H3K27me3 in EC undergoing inflammation attenuated Notch activation, endothelial inflammation, and apoptosis. Together, these findings indicate that dynamic chromatin modifications may cause an inflammatory and apoptotic switch of EC and that epigenetic reprogramming can potentially improve outcomes in endothelial inflammation-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Yash T Katakia
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Niyati P Thakkar
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Sumukh Thakar
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Ashima Sakhuja
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Raghav Goyal
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Harshita Sharma
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Rakshita Dave
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Ayushi Mandloi
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Sayan Basu
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Ishan Nigam
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Bhanu V R Kuncharam
- Department of Chemical Engineering, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Shibasish Chowdhury
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| | - Syamantak Majumder
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, India
| |
Collapse
|
10
|
Wang X, Zhu G, Wang S, Rhen J, Pang J, Zhang Z. Vessel tech: a high-accuracy pipeline for comprehensive mouse retinal vasculature characterization. Angiogenesis 2021; 24:7-11. [PMID: 33033849 PMCID: PMC7920901 DOI: 10.1007/s10456-020-09752-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 09/28/2020] [Indexed: 10/23/2022]
Abstract
Mouse retinal vasculature is a well-recognized and commonly used animal model for angiogenesis and microvascular remodeling. Morphological features of retinal vasculature reflect the vessel's biological functions, and are critical in understanding the physiological and pathological process of vascular development and disease. Here we developed a comprehensive software, Vessel Tech, using retinal vasculature images of postnatal mice. This pipeline can automatically process retinal vascular images, reconstruct vessel network with high accuracy and assess global and local vascular characteristics based on the recent machine-learning techniques. The development of Vessel Tech provides a powerful tool for vascular biologists.
Collapse
Affiliation(s)
- Xuelin Wang
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Guofu Zhu
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Shumin Wang
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Jordan Rhen
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Jinjiang Pang
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| | - Zhengwu Zhang
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| |
Collapse
|
11
|
Zhao W, Zheng Z, Aweya JJ, Wang F, Li S, Tuan TN, Yao D, Zhang Y. Litopenaeus vannamei Notch interacts with COP9 signalosome complex subunit 1 (CNS1) to negatively regulate the NF-κB pathway. J Proteomics 2020; 232:104074. [PMID: 33309928 DOI: 10.1016/j.jprot.2020.104074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 11/04/2020] [Accepted: 12/06/2020] [Indexed: 01/06/2023]
Abstract
Notch signaling pathway is a highly evolutionary conserved signaling pathway, which modulates many biological processes such as cell differentiation, tissue development and immune response. Our previous study revealed that Litopenaeus vannamei Notch (LvNotch) was involved in immune response by regulating reactive oxygen species (ROS) production in hemocytes. However, the immune regulatory networks mediated by LvNotch remain unclear in shrimp. In this study, 21 proteins that potentially interact with LvNotch were identified by GST pull-down and liquid chromatography tandem mass spectrometry (LC-MS/MS) analyses. Among these proteins, COP9 signalosome complex subunit 1 (CSN1) was chosen for further studies due to its putative role in immune response. The interaction between LvNotch and LvCSN1 was confirmed by Far-Western blot and GST pull-down analyses. In vivo knockdown of LvNotch resulted in an increase in LvCSN1 expression in hemocytes, which suggest that the COP9 signalosome complex might be negatively regulated by LvNotch. In addition, in vivo silencing of LvNotch upregulated the expression of LvDorsal, LvTNFSF and LvCrustin2 (NF-κB pathway related-genes), while their expression decreased after LvCSN1 depletion. Collectively, the current results indicate that LvNotch negatively regulates the NF-κB pathway by modulating LvCSN1 in shrimp. SIGNIFICANCE: Although the Notch signaling pathway has been implicated in the regulation of immune response in vertebrates and invertebrates, the functions and immune-related interacting networks of Notch in shrimp immune response remain unknown. In this study, twenty-one proteins including COP9 signalosome complex subunit 1 (CSN1) were identified as potential interacting partners of LvNotch. Further analysis revealed that LvNotch negatively regulates the NF-κB pathway by binding to CSN1 and modulating its expression. These findings for the first time suggest that the Notch signaling pathway has cross-talk with the NF-κB pathway in shrimp as part of the immune response.
Collapse
Affiliation(s)
- Weiling Zhao
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China
| | - Zhihong Zheng
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China
| | - Jude Juventus Aweya
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China
| | - Fan Wang
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China
| | - Shengkang Li
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China
| | - Tran Ngoc Tuan
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China
| | - Defu Yao
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China.
| | - Yueling Zhang
- Institute of Marine Sciences and Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou 515063, China; STU-UMT Joint Shellfish Research Laboratory, Shantou University, Shantou 515063, China; Southern Marine Science and Engineering Guangdong Laboratory, Guangzhou 511458, China.
| |
Collapse
|
12
|
Abstract
All organisms growing beyond the oxygen diffusion limit critically depend on a functional vasculature for survival. Yet blood vessels are far more than passive, uniform conduits for oxygen and nutrient supply. A remarkable organotypic heterogeneity is brought about by tissue-specific differentiated endothelial cells (lining the blood vessels' lumen) and allows blood vessels to deal with organ-specific demands for homeostasis. On the flip side, when blood vessels go awry, they promote life-threatening diseases characterized by endothelial cells inappropriately adopting an angiogenic state (eg, tumor vascularization) or becoming dysfunctional (eg, diabetic microvasculopathies), calling respectively for antiangiogenic therapies and proangiogenic/vascular regenerative strategies. In solid tumors, despite initial enthusiasm, growth factor-based (mostly anti-VEGF [vascular endothelial growth factor]) antiangiogenic therapies do not sufficiently live up to the expectations in terms of efficiency and patient survival, in part, due to intrinsic and acquired therapy resistance. Tumors cunningly deploy alternative growth factors than the ones targeted by the antiangiogenic therapies to reinstigate angiogenesis or revert to other ways of securing blood flow, independently of the targeted growth factors. In trying to alleviate tissue ischemia and to repair dysfunctional or damaged endothelium, local in-tissue administration of (genes encoding) proangiogenic factors or endothelial (stem) cells harnessing regenerative potential have been explored. Notwithstanding evaluation in clinical trials, these approaches are often hampered by dosing issues and limited half-life or local retention of the administered agents. Here, without intending to provide an all-encompassing historical overview, we focus on some recent advances in understanding endothelial cell behavior in health and disease and identify novel molecular players and concepts that could eventually be considered for therapeutic targeting.
Collapse
Affiliation(s)
- Guy Eelen
- From the Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Belgium (G.E., L.T., P.C.)
| | - Lucas Treps
- From the Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Belgium (G.E., L.T., P.C.)
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China (X.L., P.C.)
| | - Peter Carmeliet
- From the Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Belgium (G.E., L.T., P.C.).,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China (X.L., P.C.)
| |
Collapse
|
13
|
Yasuda D, Kobayashi D, Akahoshi N, Ohto-Nakanishi T, Yoshioka K, Takuwa Y, Mizuno S, Takahashi S, Ishii S. Lysophosphatidic acid-induced YAP/TAZ activation promotes developmental angiogenesis by repressing Notch ligand Dll4. J Clin Invest 2019; 129:4332-4349. [PMID: 31335323 DOI: 10.1172/jci121955] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Lysophosphatidic acid (LPA) is a potent lipid mediator with various biological functions mediated through six G protein-coupled receptors (GPCRs), LPA1-6. Previous studies have demonstrated that LPA-Gα12/Gα13 signaling plays an important role in embryonic vascular development. However, the responsible LPA receptors and underlying mechanisms are poorly understood. Here, we show a critical role of LPA4 and LPA6 in developmental angiogenesis. In mice, Lpa4;Lpa6 double knockout (DKO) embryos were lethal due to global vascular deficiencies, and endothelial cell (EC)-specific Lpa4;Lpa6 DKO retinas had impaired sprouting angiogenesis. Mechanistically, LPA activated the transcriptional regulators YAP and TAZ through LPA4/LPA6-mediated Gα12/Gα13-Rho-ROCK signaling in ECs. YAP/TAZ knockdown increased β-catenin- and Notch intracellular domain (NICD)-mediated endothelial expression of the Notch ligand delta-like 4 (DLL4). Fibrin gel sprouting assay revealed that LPA4/LPA6, Gα12/Gα13, or YAP/TAZ knockdown consistently blocked EC sprouting, which was rescued by a Notch inhibitor. Of note, the inhibition of Notch signaling also ameliorated impaired retinal angiogenesis in EC-specific Lpa4;Lpa6 DKO mice. Overall, these results suggest that the Gα12/Gα13-coupled receptors LPA4 and LPA6 synergistically regulate endothelial Dll4 expression through YAP/TAZ activation. This could in part account for the mechanism of YAP/TAZ-mediated developmental angiogenesis. Our findings provide a novel insight into the biology of GPCR-activated YAP/TAZ.
Collapse
Affiliation(s)
- Daisuke Yasuda
- Department of Immunology, Akita University Graduate School of Medicine, Akita, Japan
| | - Daiki Kobayashi
- Department of Immunology, Akita University Graduate School of Medicine, Akita, Japan
| | - Noriyuki Akahoshi
- Department of Immunology, Akita University Graduate School of Medicine, Akita, Japan
| | - Takayo Ohto-Nakanishi
- Department of Immunology, Akita University Graduate School of Medicine, Akita, Japan
| | - Kazuaki Yoshioka
- Department of Vascular Molecular Physiology, Kanazawa University Graduate School of Medicine, Ishikawa, Japan
| | - Yoh Takuwa
- Department of Vascular Molecular Physiology, Kanazawa University Graduate School of Medicine, Ishikawa, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center, University of Tsukuba, Ibaraki, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center, University of Tsukuba, Ibaraki, Japan
| | - Satoshi Ishii
- Department of Immunology, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
14
|
Chen W, Xia P, Wang H, Tu J, Liang X, Zhang X, Li L. The endothelial tip-stalk cell selection and shuffling during angiogenesis. J Cell Commun Signal 2019; 13:291-301. [PMID: 30903604 DOI: 10.1007/s12079-019-00511-z] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 02/25/2019] [Indexed: 12/17/2022] Open
Abstract
Angiogenesis is a critical, fine-tuned, multi-staged biological process. Tip-stalk cell selection and shuffling are the building blocks of sprouting angiogenesis. Accumulated evidences show that tip-stalk cell selection and shuffling are regulated by a variety of physical, chemical and biological factors, especially the interaction among multiple genes, their products and environments. The classic Notch-VEGFR, Slit-Robo, ECM-binding integrin, semaphorin and CCN family play important roles in tip-stalk cell selection and shuffling. In this review, we outline the progress and prospect in the mechanism and the roles of the various molecules and related signaling pathways in endothelial tip-stalk cell selection and shuffling. In the future, the regulators of tip-stalk cell selection and shuffling would be the potential markers and targets for angiogenesis.
Collapse
Affiliation(s)
- Wenqi Chen
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Peng Xia
- Department of Anesthesia, Jilin Provincial People's Hospital, Changchun, China
| | - Heping Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, China
| | - Jihao Tu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xinyue Liang
- The First Hospital of Jilin University, Changchun, China
| | - Xiaoling Zhang
- The First Hospital of Jilin University, Changchun, China. .,Institute of Immunology, Jilin University, Changchun, China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China.
| |
Collapse
|
15
|
Zhang Y, Coarfa C, Dong X, Jiang W, Hayward-Piatkovskyi B, Gleghorn JP, Lingappan K. MicroRNA-30a as a candidate underlying sex-specific differences in neonatal hyperoxic lung injury: implications for BPD. Am J Physiol Lung Cell Mol Physiol 2019; 316:L144-L156. [PMID: 30382766 PMCID: PMC6383497 DOI: 10.1152/ajplung.00372.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/22/2018] [Accepted: 10/29/2018] [Indexed: 02/07/2023] Open
Abstract
Premature male neonates are at a greater risk of developing bronchopulmonary dysplasia (BPD). The reasons underlying sexually dimorphic outcomes in premature neonates are not known. The role of miRNAs in mediating sex biases in BPD is understudied. Analysis of the pulmonary transcriptome revealed that a large percentage of angiogenesis-related differentially expressed genes are miR-30a targets. We tested the hypothesis that there is differential expression of miR-30a in vivo and in vitro in neonatal human pulmonary microvascular endothelial cells (HPMECs) upon exposure to hyperoxia. Neonatal male and female mice (C57BL/6) were exposed to hyperoxia [95% fraction of inspired oxygen (FiO2), postnatal day ( PND) 1-5] and euthanized on PND 7 and 21. HPMECs (18-24-wk gestation donors) were subjected to hyperoxia (95% O2 and 5% CO2) or normoxia (air and 5% CO2) up to 72 h. miR-30a expression was increased in both males and females in the acute phase ( PND 7) after hyperoxia exposure. However, at PND 21 (recovery phase), female mice showed significantly higher miR-30a expression in the lungs compared with male mice. Female HPMECs showed greater expression of miR-30a in vitro upon exposure to hyperoxia. Delta-like ligand 4 (Dll4) was an miR-30a target in HPMECs and showed sex-specific differential expression. miR-30a increased angiogenic sprouting in vitro in female HPMECs. Lastly, we show decreased expression of miR-30a and increased expression of DLL4 in human BPD lung samples compared with controls. These results support the hypothesis that miR-30a could, in part, contribute to the sex-specific molecular mechanisms in play that lead to the sexual dimorphism in BPD.
Collapse
Affiliation(s)
- Yuhao Zhang
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine , Houston, Texas
| | - Cristian Coarfa
- Advanced Technology Cores, Baylor College of Medicine , Houston, Texas
| | - Xiaoyu Dong
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine , Houston, Texas
| | - Weiwu Jiang
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine , Houston, Texas
| | | | - Jason P Gleghorn
- Department of Biological Sciences, University of Delaware , Newark, Delaware
- Department of Biomedical Engineering, University of Delaware , Newark, Delaware
| | - Krithika Lingappan
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine , Houston, Texas
| |
Collapse
|
16
|
Zhu G, Lin Y, Liu H, Jiang D, Singh S, Li X, Yu Z, Fan L, Wang S, Rhen J, Li W, Xu Y, Ge J, Pang J. Dll4-Notch1 signaling but not VEGF-A is essential for hyperoxia induced vessel regression in retina. Biochem Biophys Res Commun 2018; 507:400-406. [PMID: 30448061 DOI: 10.1016/j.bbrc.2018.11.051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 11/09/2018] [Indexed: 02/03/2023]
Abstract
It is well recognized that decreased vascular endothelial growth factor A (VEGF-A) mRNA plays an important role in retinal vessel regression induced by hyperoxia. However, this concept has been challenged by increasing new evidence. Furthermore, VEGF-A strongly enhances Dll4 expression and inhibition of Dll4-Notch signaling leads to excessive sprouting angiogenesis. Recently, it is shown that inactivation of Dll4-Notch1 signaling reduce hyperoxia induced vessel regression. It is unknown whether sprouting angiogenesis contributes to the protective effect or not and further investigations are needed. Moreover, the expression of Dll4 or Notch1 activation in the regressing plexus remains elucidated. To determine the role of VEGF-A and Dll4-Notch1 signaling in hyperoxia induced vascular regression in the retina, we used mice at postnatal day 5 (P5) - P7. Hyperoxia induced massive vascular regression in the central plexus but not in the angiogenic plexus and had no effect on sprouting angiogenesis. Immunostaining showed that VEGF-A was significantly repressed in the angiogenic front region after hyperoxia exposure but not detectable in the central area of both normoxia and hyperoxia treated retinas. In contrast, Notch ligand Delta-like 4 (Dll4) and Notch1 intracellular domain (N1-ICD) expression were inhibited in the regressing capillaries of central retina but comparable in the angiogenic plexus after high oxygen treatment. Moreover, administration of Dll4 neutralizing antibody or γ-Secretase inhibitor DAPT significantly aggravated vessel regression induced by short-time hyperoxia administration. Our data show that repressed Dll4-Notch1 signaling pathway but not downregulation of VEGF-A expression are responsible for hyperoxia induced pervasive vessel regression.
Collapse
Affiliation(s)
- Guofu Zhu
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China
| | - Ying Lin
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China
| | - Hao Liu
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China
| | - Dongyang Jiang
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China
| | - Shekhar Singh
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China
| | - Xiankai Li
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China
| | - Ze Yu
- College of Laboratory Science, Dalian Medical University, Dalian, China
| | - Linlin Fan
- College of Laboratory Science, Dalian Medical University, Dalian, China
| | - Shumin Wang
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USEA
| | - Jordan Rhen
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USEA
| | - Weiming Li
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China
| | - Yawei Xu
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China
| | - Junbo Ge
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China.
| | - Jinjiang Pang
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People's Hospital, Tongji University School of Medicine, China; Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USEA.
| |
Collapse
|
17
|
Appari M, Channon KM, McNeill E. Metabolic Regulation of Adipose Tissue Macrophage Function in Obesity and Diabetes. Antioxid Redox Signal 2018; 29:297-312. [PMID: 28661198 PMCID: PMC6012981 DOI: 10.1089/ars.2017.7060] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Obesity and diabetes are associated with chronic activation of inflammatory pathways that are important mechanistic links between insulin resistance (IR), type 2 diabetes (T2D), and cardiovascular disease pathogenesis. The development of these metabolic diseases is associated with changes in both the number and phenotype of adipose tissue macrophages (ATMs). Emerging lines of evidence have shown that ATMs release proinflammatory cytokines similar to classically activated M1 macrophages, which directly contribute to IR or T2D. In contrast, adipose tissue (AT) from lean healthy individuals contains macrophages with a less inflammatory M2 phenotype. Recent Advances: Recent research has shown that macrophage phenotype is linked to profound changes in macrophage cellular metabolism. CRITICAL ISSUES This review focuses on the role of macrophages in AT inflammation and obesity, and the metabolic changes in macrophage function that occur with activation that underpin their role in the pathogenesis of IR and T2D. We highlight current targets for altering macrophage metabolism from both within the field of metabolic disease and AT biology and more widely within inflammatory biology. FUTURE DIRECTIONS As our knowledge of macrophage metabolic programming in AT builds, there will be increasing scope for targeting this aspect of macrophage biology as a therapeutic strategy in metabolic diseases. Antioxid. Redox Signal. 29, 297-312.
Collapse
Affiliation(s)
- Mahesh Appari
- 1 Division of Cardiovascular Medicine, British Heart Foundation Centre for Research Excellence, John Radcliffe Hospital, University of Oxford , Oxford, United Kingdom .,2 Wellcome Trust Centre for Human Genetics, University of Oxford , Oxford, United Kingdom
| | - Keith M Channon
- 1 Division of Cardiovascular Medicine, British Heart Foundation Centre for Research Excellence, John Radcliffe Hospital, University of Oxford , Oxford, United Kingdom .,2 Wellcome Trust Centre for Human Genetics, University of Oxford , Oxford, United Kingdom
| | - Eileen McNeill
- 1 Division of Cardiovascular Medicine, British Heart Foundation Centre for Research Excellence, John Radcliffe Hospital, University of Oxford , Oxford, United Kingdom .,2 Wellcome Trust Centre for Human Genetics, University of Oxford , Oxford, United Kingdom
| |
Collapse
|
18
|
Majumder S, Thieme K, Batchu SN, Alghamdi TA, Bowskill BB, Kabir MG, Liu Y, Advani SL, White KE, Geldenhuys L, Tennankore KK, Poyah P, Siddiqi FS, Advani A. Shifts in podocyte histone H3K27me3 regulate mouse and human glomerular disease. J Clin Invest 2017; 128:483-499. [PMID: 29227285 DOI: 10.1172/jci95946] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/31/2017] [Indexed: 01/09/2023] Open
Abstract
Histone protein modifications control fate determination during normal development and dedifferentiation during disease. Here, we set out to determine the extent to which dynamic changes to histones affect the differentiated phenotype of ordinarily quiescent adult glomerular podocytes. To do this, we examined the consequences of shifting the balance of the repressive histone H3 lysine 27 trimethylation (H3K27me3) mark in podocytes. Adriamycin nephrotoxicity and subtotal nephrectomy (SNx) studies indicated that deletion of the histone methylating enzyme EZH2 from podocytes decreased H3K27me3 levels and sensitized mice to glomerular disease. H3K27me3 was enriched at the promoter region of the Notch ligand Jag1 in podocytes, and derepression of Jag1 by EZH2 inhibition or knockdown facilitated podocyte dedifferentiation. Conversely, inhibition of the Jumonji C domain-containing demethylases Jmjd3 and UTX increased the H3K27me3 content of podocytes and attenuated glomerular disease in adriamycin nephrotoxicity, SNx, and diabetes. Podocytes in glomeruli from humans with focal segmental glomerulosclerosis or diabetic nephropathy exhibited diminished H3K27me3 and heightened UTX content. Analogous to human disease, inhibition of Jmjd3 and UTX abated nephropathy progression in mice with established glomerular injury and reduced H3K27me3 levels. Together, these findings indicate that ostensibly stable chromatin modifications can be dynamically regulated in quiescent cells and that epigenetic reprogramming can improve outcomes in glomerular disease by repressing the reactivation of developmental pathways.
Collapse
Affiliation(s)
- Syamantak Majumder
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Karina Thieme
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Sri N Batchu
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Tamadher A Alghamdi
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Bridgit B Bowskill
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - M Golam Kabir
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Youan Liu
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Suzanne L Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Kathryn E White
- Electron Microscopy Research Services, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | | | - Penelope Poyah
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ferhan S Siddiqi
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Andrew Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|