1
|
Yang H, Han RY, Gong RW, Zhang YJ, Yang SS, Xu GZ, Liu W. CST3 alleviates retinal vascular leakage by regulating the Rap1 signaling pathway. Exp Eye Res 2024; 247:110042. [PMID: 39147193 DOI: 10.1016/j.exer.2024.110042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/21/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Retinal vascular leakage is a major event in several retinal diseases, including diabetic retinopathy (DR). In a previous study, we demonstrated that the aqueous humor concentration of Cystatin C (CST3), a physiological inhibitor of cysteine protease, is negatively correlated with the severity of diabetic macular edema. However, its function in the retina has not been clearly elucidated. In this study, we found a significant decrease in the aqueous humor concentration of CST3 with DR progression. Furthermore, we found that CST3 was expressed in retinal endothelial cells and that its expression was significantly downregulated in high glucose-treated human retinal microvascular endothelial cells (HRMECs) and the retinal vessels of oxygen-induced retinopathy (OIR) mice. Silencing CST3 expression resulted in decreased HRMEC migration and tubule formation ability. Exogenous addition of the CST3 protein significantly improved HRMEC migration and tubular formation. In-vivo experiments demonstrated that CST3 silencing induced retinal vascular leakage in WT mice, while its intravitreal injection significantly reduced retinal leakage in OIR mice. Mechanistically, CST3 promoted the expression of the downstream adhesion molecules, claudin5, VE-cadherin, and ZO-1, in retinal vascular cells by regulating the Rap1 signaling pathway. Therefore, this study revealed a novel mechanism by which CST3 improves retinal vascular function and provided evidence that it is a potential therapeutic target for retinal vascular leakage.
Collapse
Affiliation(s)
- Hong Yang
- Department of Ophthalmology, Eye, ENT Hospital of Fudan University, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment, Restoration, Fudan University, Shanghai, 200031, China
| | - Ru-Yi Han
- Department of Ophthalmology, Eye, ENT Hospital of Fudan University, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment, Restoration, Fudan University, Shanghai, 200031, China
| | - Ruo-Wen Gong
- Department of Ophthalmology, Eye, ENT Hospital of Fudan University, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment, Restoration, Fudan University, Shanghai, 200031, China
| | - Ya-Juan Zhang
- Sixth Affiliated Hospital of Kunming Medical University, Yun Nan, 653100, China
| | - Shi-Shi Yang
- Sixth Affiliated Hospital of Kunming Medical University, Yun Nan, 653100, China
| | - Ge-Zhi Xu
- Department of Ophthalmology, Eye, ENT Hospital of Fudan University, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment, Restoration, Fudan University, Shanghai, 200031, China.
| | - Wei Liu
- Department of Ophthalmology, Eye, ENT Hospital of Fudan University, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment, Restoration, Fudan University, Shanghai, 200031, China.
| |
Collapse
|
2
|
Ren Y, Liang H, Xie M, Zhang M. Natural plant medications for the treatment of retinal diseases: The blood-retinal barrier as a clue. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155568. [PMID: 38795692 DOI: 10.1016/j.phymed.2024.155568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/15/2024] [Accepted: 03/23/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND Retinal diseases significantly contribute to the global burden of visual impairment and blindness. The occurrence of retinal diseases is often accompanied by destruction of the blood‒retinal barrier, a vital physiological structure responsible for maintaining the stability of the retinal microenvironment. However, detailed summaries of the factors damage the blood‒retinal barrier and treatment methods involving natural plant medications are lacking. PURPOSE To comprehensively summarize and analyze the protective effects of active substances in natural plant medications on damage to the blood-retina barrier that occurs when retinal illnesses, particularly diabetic retinopathy, and examine their medicinal value and future development prospects. METHODS In this study, we searched for studies published in the ScienceDirect, PubMed, and Web of Science databases. The keywords used included natural plant medications, plants, natural herbs, blood retinal barrier, retinal diseases, diabetic retinopathy, age-related macular degeneration, and uveitis. Chinese herbal compound articles, non-English articles, warning journals, and duplicates were excluded from the analysis. RESULTS The blood‒retinal barrier is susceptible to high glucose, aging, immune responses, and other factors that destroy retinal homeostasis, resulting in pathological changes such as apoptosis and increased vascular permeability. Existing studies have shown that the active compounds or extracts of many natural plants have the effect of repairing blood-retinal barrier dysfunction. Notably, berberine, puerarin, and Lycium barbarum polysaccharides exhibited remarkable therapeutic effects. Additionally, curcumin, astragaloside IV, hesperidin, resveratrol, ginsenoside Rb1, luteolin, and Panax notoginseng saponins can effectively protect the blood‒retinal barrier by interfering with distinct pathways. The active ingredients found in natural plant medications primarily repair the blood‒retinal barrier by modulating pathological factors such as oxidative stress, inflammation, pyroptosis, and autophagy, thereby alleviating retinal diseases. CONCLUSION This review summarizes a series of plant extracts and plant active compounds that can treat retinal diseases by preventing and treating blood‒retinal barrier damage and provides reference for the research of new drugs for treating retinal diseases.
Collapse
Affiliation(s)
- Yuan Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Huan Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Mengjun Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| | - Mei Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| |
Collapse
|
3
|
Roy S, Roy S, Halder S, Jana K, Ukil A. Leishmania exploits host cAMP/EPAC/calcineurin signaling to induce an IL-33-mediated anti-inflammatory environment for the establishment of infection. J Biol Chem 2024; 300:107366. [PMID: 38750790 PMCID: PMC11208913 DOI: 10.1016/j.jbc.2024.107366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 06/10/2024] Open
Abstract
Host anti-inflammatory responses are critical for the progression of visceral leishmaniasis, and the pleiotropic cytokine interleukin (IL)-33 was found to be upregulated in infection. Here, we documented that IL-33 induction is a consequence of elevated cAMP-mediated exchange protein activated by cAMP (EPAC)/calcineurin-dependent signaling and essential for the sustenance of infection. Leishmania donovani-infected macrophages showed upregulation of IL-33 and its neutralization resulted in decreased parasite survival and increased inflammatory responses. Infection-induced cAMP was involved in IL-33 production and of its downstream effectors PKA and EPAC, only the latter was responsible for elevated IL-33 level. EPAC initiated Rap-dependent phospholipase C activation, which triggered the release of intracellular calcium followed by calcium/calmodulin complex formation. Screening of calmodulin-dependent enzymes affirmed involvement of the phosphatase calcineurin in cAMP/EPAC/calcium/calmodulin signaling-induced IL-33 production and parasite survival. Activated calcineurin ensured nuclear localization of the transcription factors, nuclear factor of activated T cell 1 and hypoxia-inducible factor 1 alpha required for IL-33 transcription, and we further confirmed this by chromatin immunoprecipitation assay. Administering specific inhibitors of nuclear factor of activated T cell 1 and hypoxia-inducible factor 1 alpha in BALB/c mouse model of visceral leishmaniasis decreased liver and spleen parasite burden along with reduction in IL-33 level. Splenocyte supernatants of inhibitor-treated infected mice further documented an increase in tumor necrosis factor alpha and IL-12 level with simultaneous decrease of IL-10, thereby indicating an overall disease-escalating effect of IL-33. Thus, this study demonstrates that cAMP/EPAC/calcineurin signaling is crucial for the activation of IL-33 and in effect creates anti-inflammatory responses, essential for infection.
Collapse
Affiliation(s)
- Souravi Roy
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | - Shalini Roy
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | - Satyajit Halder
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Kuladip Jana
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Anindita Ukil
- Department of Biochemistry, University of Calcutta, Kolkata, India.
| |
Collapse
|
4
|
Wu J, Wang H, Wang N, Wang Z, Zhu Q. TIE2 expression in hypertensive ICH and its therapeutic modulation with AKB-9778: Implications for brain vascular health. Exp Neurol 2024; 374:114685. [PMID: 38195021 DOI: 10.1016/j.expneurol.2024.114685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/27/2023] [Accepted: 01/05/2024] [Indexed: 01/11/2024]
Abstract
Hypertensive intracerebral hemorrhage (ICH) is a devastating condition, the molecular underpinnings of which remain not fully understood. By leveraging high-throughput transcriptome sequencing and network pharmacology analysis, this study unveils the significant role of the tyrosine kinase with immunoglobulin-like and EGF-like domains 2 (TIE2) in ICH pathogenesis. Compared to controls, a conspicuous downregulation of TIE2 was observed in the cerebral blood vessels of hypertensive ICH mice. In vitro assays with human brain microvascular endothelial cells (HBMEC), HBEC-5i revealed that modulation of TIE2 expression significantly influences cellular proliferation, migration, and angiogenesis, mediated via the Rap1/MEK/ERK signaling pathway. Notably, the small molecule AKB-9778 was identified to target and activate TIE2, affecting the functional attributes of HBEC-5i. In vivo experiments further demonstrated that combining AKB-9778 with antihypertensive drugs could mitigate the incidence and volume of bleeding in hypertensive ICH mouse models, suggesting potential therapeutic implications.
Collapse
Affiliation(s)
- Jingkun Wu
- Department of Neurosurgery, Affiliated Hospital of Hebei Engineering University, Handan 056002, PR China
| | - Hongbin Wang
- Department of Neurosurgery, Affiliated Hospital of Hebei Engineering University, Handan 056002, PR China
| | - Naizhu Wang
- Department of Neurosurgery, Affiliated Hospital of Hebei Engineering University, Handan 056002, PR China
| | - Zai Wang
- Science and Education Division, Affiliated Hospital of Hebei Engineering University, Handan 056002, PR China
| | - Qinghua Zhu
- Department of Neurosurgery, Affiliated Hospital of Hebei Engineering University, Handan 056002, PR China.
| |
Collapse
|
5
|
Li Z, Liu Q, Cai Y, Ye N, He Z, Yao Y, Ding Y, Wang P, Qi C, Zheng L, Wang L, Zhou J, Zhang QQ. EPAC inhibitor suppresses angiogenesis and tumor growth of triple-negative breast cancer. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167114. [PMID: 38447883 DOI: 10.1016/j.bbadis.2024.167114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/08/2024]
Abstract
AIMS Exchange protein directly activated by cAMP 1 (EPAC1), a major isoform of guanine nucleotide exchange factors, is highly expressed in vascular endothelia cells and regulates angiogenesis in the retina. High intratumor microvascular densities (MVD) resulting from angiogenesis is responsible for breast cancer development. Downregulation of EPAC1 in tumor cell reduces triple-negative breast cancer (TNBC)-induced angiogenesis. However, whether Epac1 expressed in vascular endothelial cells contributes to angiogenesis and tumor development of TNBC remains elusive. MAIN METHODS We employed NY0123, a previously identified potent EPAC inhibitor, to explore the anti-angiogenic biological role of EPAC1 in vitro and in vivo through vascular endothelial cells, rat aortic ring, Matrigel plug, and chick embryo chorioallantoic membrane (CAM) and yolk sac membrane (YSM) assays, as well as the in vivo xenograft tumor models of TNBC in both chick embryo and mice. KEY FINDINGS Inhibiting EPAC1 in vascular endothelial cells by NY0123 significantly suppresses angiogenesis and tumor growth of TNBC. In addition, NY0123 possesses a better inhibitory efficacy than ESI-09, a reported specific EPAC inhibitor tool compound. Importantly, inhibiting EPAC1 in vascular endothelia cells regulates the typical angiogenic signaling network, which is associated with not only vascular endothelial growth factor (VEGF)/vascular endothelial growth factor receptor-2 (VEGFR2) signaling, but also PI3K/AKT, MEK/ERK and Notch pathway. CONCLUSIONS Our findings support that EPAC1 may serve as an effective anti-angiogenic therapeutic target of TNBC, and EPAC inhibitor NY0123 has the therapeutic potential to be developed for the treatment of TNBC.
Collapse
Affiliation(s)
- Zishuo Li
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qiao Liu
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yuhao Cai
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Na Ye
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Zinan He
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yuying Yao
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yi Ding
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Pingyuan Wang
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Cuiling Qi
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lingyun Zheng
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lijing Wang
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States.
| | - Qian-Qian Zhang
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
6
|
Fu Q, Wang Y, Yan C, Xiang YK. Phosphodiesterase in heart and vessels: from physiology to diseases. Physiol Rev 2024; 104:765-834. [PMID: 37971403 PMCID: PMC11281825 DOI: 10.1152/physrev.00015.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/17/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
Phosphodiesterases (PDEs) are a superfamily of enzymes that hydrolyze cyclic nucleotides, including cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Both cyclic nucleotides are critical secondary messengers in the neurohormonal regulation in the cardiovascular system. PDEs precisely control spatiotemporal subcellular distribution of cyclic nucleotides in a cell- and tissue-specific manner, playing critical roles in physiological responses to hormone stimulation in the heart and vessels. Dysregulation of PDEs has been linked to the development of several cardiovascular diseases, such as hypertension, aneurysm, atherosclerosis, arrhythmia, and heart failure. Targeting these enzymes has been proven effective in treating cardiovascular diseases and is an attractive and promising strategy for the development of new drugs. In this review, we discuss the current understanding of the complex regulation of PDE isoforms in cardiovascular function, highlighting the divergent and even opposing roles of PDE isoforms in different pathogenesis.
Collapse
Affiliation(s)
- Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Ying Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Chen Yan
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York, United States
| | - Yang K Xiang
- Department of Pharmacology, University of California at Davis, Davis, California, United States
- Department of Veterans Affairs Northern California Healthcare System, Mather, California, United States
| |
Collapse
|
7
|
Chen X, Su D, Sun Z, Fu Y, Hu Y, Zhang Y, Zhang X, Wei Q, Zhu W, Ma X, Hu S. Preliminary study on whole genome methylation and transcriptomics in age-related cataracts. Gene 2024; 898:148096. [PMID: 38128790 DOI: 10.1016/j.gene.2023.148096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/10/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
DNA methylation plays an important role in the occurrence and development of age-related cataracts (ARC). This study aims to reveal potential epigenetic biomarkers of ARC by detecting modifications to the DNA methylation patterns of genes shown to be related to ARC by transcriptomics. The MethylationEPIC BeadChip (850 K) was used to analyze the DNA methylation levels in ARC patients and unaffected controls, and the Pearson correlation test was used to perform genome-wide integration analysis of DNA methylation and transcriptome data. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases were used to perform functional analysis of the whole genome, promoter regions (TSS1500/TSS200), and the associated differentially methylated genes (DMG). Pyrosequencing was used to verify the methylation levels of the selected genes. The results showed that, compared with the control group, a total of 52,705 differentially methylated sites were detected in the ARC group, of which 13,858 were hypermethylated and 38,847 were hypomethylated. GO and KEGG analyses identified functions related to the cell membrane, the calcium signaling pathway, and their possible molecular mechanisms. Then, 57 DMGs with negative promoter methylation correlations were screened by association analysis. Pyrosequencing verified that the ARC group had higher methylation levels of C3 and CCKAR and lower methylation levels of NLRP3, LEFTY1, and GPR35 compared with the control group. In summary, our study reveals the whole-genome DNA methylation patterns and gene expression profiles in ARC, and the molecular markers of methylation identified herein may aid in the prevention, diagnosis, treatment, and prognosis of ARC.
Collapse
Affiliation(s)
- Xiaoya Chen
- Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang, China
| | - Dongmei Su
- Department of Genetics, NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning, Health Department, Beijing 100081, China; Graduate School, Peking Union Medical College, Beijing 100081, China
| | - Zhaoyi Sun
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang, China
| | - Yanjiang Fu
- Daqing Eye Hospital, Daqing 163000, Heilongjiang, China
| | - Yuzhu Hu
- Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang, China
| | - Yue Zhang
- Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang, China
| | - Xiao Zhang
- Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang, China
| | - Qianqiu Wei
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang, China
| | - Wenna Zhu
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang, China
| | - Xu Ma
- Department of Genetics, NHC Key Laboratory of Reproductive Health Engineering Technology Research, National Research Institute for Family Planning, Health Department, Beijing 100081, China; Graduate School, Peking Union Medical College, Beijing 100081, China.
| | - Shanshan Hu
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang 157011, Heilongjiang, China.
| |
Collapse
|
8
|
Watanabe-Takano H, Kato K, Oguri-Nakamura E, Ishii T, Kobayashi K, Murata T, Tsujikawa K, Miyata T, Kubota Y, Hanada Y, Nishiyama K, Watabe T, Fässler R, Ishii H, Mochizuki N, Fukuhara S. Endothelial cells regulate alveolar morphogenesis by constructing basement membranes acting as a scaffold for myofibroblasts. Nat Commun 2024; 15:1622. [PMID: 38438343 PMCID: PMC10912381 DOI: 10.1038/s41467-024-45910-y] [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: 04/17/2023] [Accepted: 02/06/2024] [Indexed: 03/06/2024] Open
Abstract
Alveologenesis is a spatially coordinated morphogenetic event, during which alveolar myofibroblasts surround the terminal sacs constructed by epithelial cells and endothelial cells (ECs), then contract to form secondary septa to generate alveoli in the lungs. Recent studies have demonstrated the important role of alveolar ECs in this morphogenetic event. However, the mechanisms underlying EC-mediated alveologenesis remain unknown. Herein, we show that ECs regulate alveologenesis by constructing basement membranes (BMs) acting as a scaffold for myofibroblasts to induce septa formation through activating mechanical signaling. Rap1, a small GTPase of the Ras superfamily, is known to stimulate integrin-mediated cell adhesions. EC-specific Rap1-deficient (Rap1iECKO) mice exhibit impaired septa formation and hypo-alveolarization due to the decreased mechanical signaling in myofibroblasts. In Rap1iECKO mice, ECs fail to stimulate integrin β1 to recruit Collagen type IV (Col-4) into BMs required for myofibroblast-mediated septa formation. Consistently, EC-specific integrin β1-deficient mice show hypo-alveolarization, defective mechanical signaling in myofibroblasts, and disorganized BMs. These data demonstrate that alveolar ECs promote integrin β1-mediated Col-4 recruitment in a Rap1-dependent manner, thereby constructing BMs acting as a scaffold for myofibroblasts to induce mechanical signal-mediated alveologenesis. Thus, this study unveils a mechanism of organ morphogenesis mediated by ECs through intrinsic functions.
Collapse
Affiliation(s)
- Haruko Watanabe-Takano
- Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan.
| | - Katsuhiro Kato
- Department of Cardiology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Eri Oguri-Nakamura
- Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Tomohiro Ishii
- Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Koji Kobayashi
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Takahisa Murata
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Koichiro Tsujikawa
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Takaki Miyata
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Yoshiaki Kubota
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| | - Yasuyuki Hanada
- Department of Cardiology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
- Laboratory for Vascular and Cellular Dynamics, Department of Medical Sciences, University of Miyazaki, Miyazaki City, Miyazaki, 889-1962, Japan
| | - Koichi Nishiyama
- Laboratory for Vascular and Cellular Dynamics, Department of Medical Sciences, University of Miyazaki, Miyazaki City, Miyazaki, 889-1962, Japan
| | - Tetsuro Watabe
- Department of Biochemistry, Graduate, School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, 113-8549, Japan
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152, Martinsried, Germany
| | - Hirotaka Ishii
- Department of Anatomy and Neurobiology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-shimmachi, Suita, Osaka, 564-8565, Japan
| | - Shigetomo Fukuhara
- Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602, Japan.
| |
Collapse
|
9
|
Wang W, Li G, Ma J, Fan X, Lu J, Sun Q, Yao J, He Q. Microvascular rarefaction caused by the NOTCH signaling pathway is a key cause of TKI-apatinib-induced hypertension and cardiac damage. Front Pharmacol 2024; 15:1346905. [PMID: 38405666 PMCID: PMC10885812 DOI: 10.3389/fphar.2024.1346905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/30/2024] [Indexed: 02/27/2024] Open
Abstract
With the advancement of tumour-targeted therapy technology, the survival of cancer patients has continued to increase, and cardiovascular events have gradually become an important cause of death in cancer patients. This phenomenon occurs due to adverse cardiovascular reactions caused by the cardiovascular toxicity of antitumour therapy. Moreover, the increase in the proportion of elderly patients with cancer and cardiovascular diseases is due to the extension of life expectancy. Hypertension is the most common cardiovascular side effect of small molecule tyrosine kinase inhibitors (TKIs). The increase in blood pressure induced by TKIs and subsequent cardiovascular complications and events affect the survival and quality of life of patients and partly offset the benefits of antitumour therapy. Many studies have confirmed that in the pathogenesis of hypertension, arterioles and capillary thinness are involved in its occurrence and development. Our previous findings showing that apatinib causes microcirculation rarefaction of the superior mesenteric artery and impaired microvascular growth may inspire new therapeutic strategies for treating hypertension. Thus, by restoring microvascular development and branching patterns, total peripheral resistance and blood pressure are reduced. Therefore, exploring the key molecular targets of TKIs that inhibit the expression of angiogenic factors and elucidating the specific molecular mechanism involved are key scientific avenues for effectively promoting endothelial cell angiogenesis and achieving accurate repair of microcirculation injury in hypertension patients.
Collapse
Affiliation(s)
- WenJuan Wang
- Department of Cardiovascular Center, The First People’s Hospital of Huzhou City, Huzhou, China
| | - Guodong Li
- Department of Cardiovascular Center, The First People’s Hospital of Huzhou City, Huzhou, China
| | - Jie Ma
- Department of Hypertension Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Xin Fan
- Department of Hypertension Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Jianzhong Lu
- Department of Cardiovascular Center, The First People’s Hospital of Huzhou City, Huzhou, China
| | - Qiyin Sun
- Department of Cardiovascular Center, The First People’s Hospital of Huzhou City, Huzhou, China
| | - Jiafang Yao
- Department of Cardiovascular Center, The First People’s Hospital of Huzhou City, Huzhou, China
| | - Qingjian He
- Department of Breast and Thyroid Surgery, The First People’s Hospital of Huzhou City, Huzhou, China
| |
Collapse
|
10
|
Nakamura N, Honjo M, Yamagishi R, Sakata R, Watanabe S, Aihara M. Synergic effects of EP2 and FP receptors co-activation on Blood-Retinal Barrier and Microglia. Exp Eye Res 2023; 237:109691. [PMID: 37884204 DOI: 10.1016/j.exer.2023.109691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/25/2023] [Accepted: 10/22/2023] [Indexed: 10/28/2023]
Abstract
Macular edema (ME) is caused with disruption of the blood-retinal barrier (BRB) followed by fluid accumulation in the subretinal space. Main components of the outer and inner BRB are retinal pigment epithelial (RPE) cells and retinal microvascular endothelial cells, respectively. In addition, glial cells also participate in the functional regulation of the BRB as the member of 'neurovascular unit'. Under various stresses, cells in neurovascular units secrete inflammatory cytokines. Neuroinflammation induced by these cytokines can cause BRB dysfunction by degrading barrier-related proteins and contribute to the pathophysiology of ME. Prostaglandins (PGs) are crucial lipid mediators involved in neuroinflammation. Among PGs, a novel EP2 agonist, omidenepag (OMD) acts on not only the uveoscleral pathway but also the conventional pathway, unlike F prostanoid (FP) receptor agonists. Moreover, the combination use of the EP and the FP agonist is not recommended because of the risk of inflammation. In this study, we investigated effects of OMD and latanoprost acid (LTA), a FP agonist, on BRB and microglia in vitro and in vivo. To investigate the function of outer/inner BRB and microglia, in vitro, ARPE-19 cells, human retinal microvascular endothelial cells (HRMECs), and MG5 cells were used. Cell viability, inflammatory cytokines mRNA and protein levels, barrier morphology/function, and microglial activation were evaluated using proliferation assays, qRT-PCR, ELISA, immunocytochemistry, trans-epithelial electrical resistance, and permeability assay. Moreover, after vitreous injection into the mouse, outer BRB morphology, glial activation, and cytokine expression were assessed. Each OMD and LTA alone did not affect the viability or cytokines expression of the three types of cells. In ARPE-19 cells, the co-stimulation of OMD and LTA increased the mRNA and protein levels of inflammatory cytokines (IL-6, TNF-α, and VEGF-A) and decreased the barrier function and the junction-related protein (ZO-1 and β-catenin). By contrast in HRMECs, the co-stimulation affected significant differences in the mRNA levels of some cytokine (IL-6 and TNF-α) but enhanced the barrier function. In MG5 cells, the cytokines mRNA and size of Iba1-expressed cell were increased. A non-steroidal anti-inflammatory inhibited the barrier dysfunction and the junction-related protein downregulation in ARPE-19 cells and activation of MG5 cells. Also in vivo, the co-stimulation induced outer BRB disruption, cytokine increase, and retinal glial activation. Therefore, the co-stimulation of EP2 and FP induced the inflammatory cytokine-mediated outer BRB disruption, the enhanced inner BRB function, and the microglial activation. The BRB imbalance and the intrinsic prostaglandin production may be involved in OMD-related inflammation.
Collapse
Affiliation(s)
- Natsuko Nakamura
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Division of Vision Research, National Institute of Sensory Organs, NHO Tokyo Medical Center, Tokyo, Japan
| | - Megumi Honjo
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Reiko Yamagishi
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Rei Sakata
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sumiko Watanabe
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Makoto Aihara
- Department of Ophthalmology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
Vielmuth F, Radeva MY, Yeruva S, Sigmund AM, Waschke J. cAMP: A master regulator of cadherin-mediated binding in endothelium, epithelium and myocardium. Acta Physiol (Oxf) 2023; 238:e14006. [PMID: 37243909 DOI: 10.1111/apha.14006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/05/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
Regulation of cadherin-mediated cell adhesion is crucial not only for maintaining tissue integrity and barrier function in the endothelium and epithelium but also for electromechanical coupling within the myocardium. Therefore, loss of cadherin-mediated adhesion causes various disorders, including vascular inflammation and desmosome-related diseases such as the autoimmune blistering skin dermatosis pemphigus and arrhythmogenic cardiomyopathy. Mechanisms regulating cadherin-mediated binding contribute to the pathogenesis of diseases and may also be used as therapeutic targets. Over the last 30 years, cyclic adenosine 3',5'-monophosphate (cAMP) has emerged as one of the master regulators of cell adhesion in endothelium and, more recently, also in epithelial cells as well as in cardiomyocytes. A broad spectrum of experimental models from vascular physiology and cell biology applied by different generations of researchers provided evidence that not only cadherins of endothelial adherens junctions (AJ) but also desmosomal contacts in keratinocytes and the cardiomyocyte intercalated discs are central targets in this scenario. The molecular mechanisms involve protein kinase A- and exchange protein directly activated by cAMP-mediated regulation of Rho family GTPases and S665 phosphorylation of the AJ and desmosome adaptor protein plakoglobin. In line with this, phosphodiesterase 4 inhibitors such as apremilast have been proposed as a therapeutic strategy to stabilize cadherin-mediated adhesion in pemphigus and may also be effective to treat other disorders where cadherin-mediated binding is compromised.
Collapse
Affiliation(s)
- Franziska Vielmuth
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Mariya Y Radeva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Sunil Yeruva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Anna M Sigmund
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| |
Collapse
|
12
|
DiGuilio KM, Rybakovsky E, Baek Y, Valenzano MC, Mullin JM. The multiphasic TNF-α-induced compromise of Calu-3 airway epithelial barrier function. Exp Lung Res 2023; 49:72-85. [PMID: 37000123 DOI: 10.1080/01902148.2023.2193637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 03/09/2023] [Indexed: 04/01/2023]
Abstract
Purpose: Airway epithelial barrier leak and the involvement of proinflammatory cytokines play a key role in a variety of diseases. This study evaluates barrier compromise by the inflammatory mediator Tumor Necrosis Factor-α (TNF-α) in the human airway epithelial Calu-3 model. Methods: We examined the effects of TNF-α on barrier function in Calu-3 cell layers using Transepithelial Electrical Resistance (TER) and transepithelial diffusion of radiolabeled probe molecules. Western immunoblot analyses of tight junctional (TJ) proteins in detergent soluble fractions were performed. Results: TNF-α dramatically reduced TER and increased paracellular permeability of both 14C-D-mannitol and the larger 5 kDa probe, 14C-inulin. A time course of the effects shows two separate actions on barrier function. An initial compromise of barrier function occurs 2-4 hours after TNF-α exposure, followed by complete recovery of barrier function by 24 hrs. Beginning 48 hrs. post-exposure, a second more sustained barrier compromise ensues, in which leakiness persists through 144 hrs. There were no changes in TJ proteins observed at 3 hrs. post exposure, but significant increases in claudins-2, -3, -4, and -5, as well as a decrease in occludin were seen at 72 hrs. post TNF-α exposure. Both the 2-4 hr. and the 72 hr. TNF-α induced leaks are shown to be mediated by the ERK signaling pathway. Conclusion: TNF-α induced a multiphasic transepithelial leak in Calu-3 cell layers that was shown to be ERK mediated, as well as involve changes in the TJ complex. The micronutrients, retinoic acid and calcitriol, were effective at reducing this barrier compromise caused by TNF-α. The significance of these results for airway disease and for COVID-19 specifically are discussed.
Collapse
Affiliation(s)
| | | | - Yoongyeong Baek
- Department of Chemistry, Drexel University, Philadelphia, PA, USA
| | | | - James M Mullin
- Lankenau Institute for Medical Research, Wynnewood, PA, USA
| |
Collapse
|
13
|
Lu Y, Chai Y, Qiu J, Zhang J, Wu M, Fu Z, Wang Y, Qin C. Integrated omics analysis reveals the epigenetic mechanism of visceral hypersensitivity in IBS-D. Front Pharmacol 2023; 14:1062630. [PMID: 37007011 PMCID: PMC10064328 DOI: 10.3389/fphar.2023.1062630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Background and objective: IBS-D is a common functional bowel disease with complex etiology and without biomarker. The pathological and physiological basis of IBS-D focuses on visceral hypersensitivity. However, its epigenetic mechanism remains elusive. Our study aimed to integrate the relationship between differentially expressed miRNAs, mRNAs and proteins in IBS-D patients in order to reveal epigenetic mechanism of visceral hypersensitivity from transcription and protein levels and provide the molecular basis for discovering biomarkers of IBS-D.Methods: The intestinal biopsies from IBS-D patients and healthy volunteers were obtained for high-throughput sequencing of miRNAs and mRNAs. The differential miRNAs were selected and verified by q-PCR experiment followed by target mRNA prediction. Biological functions were respectively analyzed for target mRNAs, differential mRNAs and the previously identified differential proteins in order to explore the characteristic involved visceral hypersensitivity. At last, interaction analysis of miRNAs, mRNAs and proteins was performed for the epigenetic regulation mechanism from transcription and protein levels.Results: Thirty-three miRNAs were found to be differentially expressed in IBS-D and five of them were further confirmed, including upregulated hsa-miR-641, hsa-miR-1843, hsa-let-7d-3p and downregulated hsa-miR-219a-5p, hsa-miR-19b-1-5p. In addition, 3,812 differential mRNAs were identified. Thirty intersecting molecules were found from the analysis on the target mRNAs of miRNAs and mRNAs. Fourteen intersecting molecules were obtained from the analysis on the target mRNAs and proteins, and thirty-six intersecting molecules were identified from analysis on the proteins and different mRNAs. According to the integrated analysis of miRNA-mRNA-protein, we noticed two new molecules COPS2 regulated by hsa-miR-19b-1-5p and MARCKS regulated by hsa-miR-641. Meanwhile some critical signaling pathways in IBS-D were found such as MAPK, GABAergic synapse, Glutamatergic synapse, and Adherens junction.Conclusion: The expressions of hsa-miR-641, hsa-miR-1843, hsa-let-7d-3p, hsa-miR-219a-5p, and hsa-miR-19b-1-5p in the intestinal tissues of IBS-D patients were significantly different. Moreover, they could regulate a variety of molecules and signaling pathways, which were involved in the multifaceted and multilevel mechanism of visceral hypersensitivity of IBS-D.
Collapse
Affiliation(s)
- Yaoyao Lu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuna Chai
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- *Correspondence: Yuna Chai, ; Yongfu Wang, ; Chongzhen Qin,
| | - Jianli Qiu
- Department of Pediatrics, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jingmin Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Menglin Wu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhe Fu
- Department of General Pediatric Surgery, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yongfu Wang
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- *Correspondence: Yuna Chai, ; Yongfu Wang, ; Chongzhen Qin,
| | - Chongzhen Qin
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- *Correspondence: Yuna Chai, ; Yongfu Wang, ; Chongzhen Qin,
| |
Collapse
|
14
|
Sun X, Wang Y, Zhao Y, Xu X, Lu W, Li Y, Bian F, Xiang L, Zhou L. Activation of the Epac/Rap1 signaling pathway alleviates blood-brain barrier disruption and brain damage following cerebral ischemia/reperfusion injury. Int Immunopharmacol 2023; 117:110014. [PMID: 36931001 DOI: 10.1016/j.intimp.2023.110014] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023]
Abstract
Blood brain barrier (BBB) destruction plays a key role in ischemia stroke, including promoting BBB leakage and brain edema, and leads to unfavorable patient prognosis. Epac/Rap1 signaling pathway is important in mediating endothelial cell barrier function. This study will investigate the regulatory role of Epac/Rap1 signaling pathway in BBB disruption after cerebral ischemia/reperfusion (CI/R) injury. CI/R model was induced by 90 min of transient middle cerebral artery occlusion (MCAO) in male C57BL/6J mice. Injection of Epac/Rap1 signaling pathway agonist was performed half an hour before the MCAO operation. The results showed that CI/R injured the tight connection of BBB and evoked the suppression of the Epac/Rap1 signaling pathway. Based on Epac activation with a cAMP analogue, 8-CPT could improve BBB disfunction by increasing the expression of tight junction protein and reducing the formation of stress fibers. In addition, 8-CPT could ameliorate neurobehavioral disorders, cerebral edema, and cerebral infarction volume in MCAO mice. Moreover, inhibition of Epac pathway with Rap1 inhibitor GGTI298 and Rac1 inhibitor NSC23766 could aggravate the damage of BBB and cerebral injury accordingly. Our results indicate that, the activation of Epac/Rap1 signaling pathway has neuroprotective effects on CI/R damaged brain, through the recovery of BBB.
Collapse
Affiliation(s)
- Xuemei Sun
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen 518055, China; The Third Affiliated Hospital of Soochow University, The First People's Hospital of Changzhou, 213000, China
| | - Yingnan Wang
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen 518055, China
| | - Yuchen Zhao
- Department of Mathematics, University of California, Los Angeles, CA 90095, USA
| | - Xinyi Xu
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen 518055, China
| | - Wenjie Lu
- Department of Pharmacology, School of Basic Medical Science, Anhui Medical University, Hefei 230032, China
| | - Yuying Li
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen 518055, China
| | - Fei Bian
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen 518055, China
| | - Lan Xiang
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen 518055, China
| | - Lanlan Zhou
- School of Medical Technology and Nursing, Shenzhen Polytechnic, Shenzhen 518055, China.
| |
Collapse
|
15
|
Abdelrahman AA, Bunch KL, Sandow PV, Cheng PNM, Caldwell RB, Caldwell RW. Systemic Administration of Pegylated Arginase-1 Attenuates the Progression of Diabetic Retinopathy. Cells 2022; 11:cells11182890. [PMID: 36139465 PMCID: PMC9497170 DOI: 10.3390/cells11182890] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 12/02/2022] Open
Abstract
Diabetic retinopathy (DR) is a serious complication of diabetes that results from sustained hyperglycemia, hyperlipidemia, and oxidative stress. Under these conditions, inducible nitric oxide synthase (iNOS) expression is upregulated in the macrophages (MΦ) and microglia, resulting in increased production of reactive oxygen species (ROS) and inflammatory cytokines, which contribute to disease progression. Arginase 1 (Arg1) is a ureohydrolase that competes with iNOS for their common substrate, L-arginine. We hypothesized that the administration of a stable form of Arg1 would deplete L-arginine’s availability for iNOS, thus decreasing inflammation and oxidative stress in the retina. Using an obese Type 2 diabetic (T2DM) db/db mouse, this study characterized DR in this model and determined if systemic treatment with pegylated Arg1 (PEG-Arg1) altered the progression of DR. PEG-Arg1 treatment of db/db mice thrice weekly for two weeks improved visual function compared with untreated db/db controls. Retinal expression of inflammatory factors (iNOS, IL-1β, TNF-α, IL-6) was significantly increased in the untreated db/db mice compared with the lean littermate controls. The increased retinal inflammatory and oxidative stress markers in db/db mice were suppressed with PEG-Arg1 treatment. Additionally, PEG-Arg1 treatment restored the blood–retinal barrier (BRB) function, as evidenced by the decreased tissue albumin extravasation and an improved endothelial ZO-1 tight junction integrity compared with untreated db/db mice.
Collapse
Affiliation(s)
- Ammar A. Abdelrahman
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Katharine L. Bunch
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Porsche V. Sandow
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Paul N-M Cheng
- Bio-Cancer Treatment International, Bioinformatics Building, Hong Kong Science Park, Tai Po, Hong Kong SAR 511513, China
| | - Ruth B. Caldwell
- Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Department of Cell Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - R. William Caldwell
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Correspondence: ; Tel.: +1-706-721-2345
| |
Collapse
|
16
|
Liu C, Ren Y, Sui X, Liu K, Zhang J, Wang Z, Li Y, Zhang Y. Integrating network pharmacology, transcriptomics, and molecular simulation to reveal the mechanism of tert-butylhydroquinone for treating diabetic retinopathy. Eur J Pharmacol 2022; 931:175215. [PMID: 35987258 DOI: 10.1016/j.ejphar.2022.175215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 08/05/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022]
Abstract
Diabetic retinopathy (DR), a common microvascular complication of diabetes mellitus, is a significant cause of blindness. As one of the crucial factors in the pathogenesis of DR, oxidative stress provides new insights for the treatment of DR. Tert-butylhydroquinone (TBHQ), an efficient phenolic antioxidant, has been proved to inhibit diabetic retina injury. However, the mechanism of TBHQ for DR treatment is still unclear. The present study was designed to investigate the potential mechanism of TBHQ for treating DR. Firstly, the potential targets of TBHQ and DR were selected to construct protein-protein interaction network, which was applied to illustrate the potential mechanism of TBHQ against DR. Combined with transcriptomics and molecular simulation, the potential mechanisms were systematically verified. The results showed that TBHQ inhibited retinal microvascular injury by regulating oxidative stress, inflammation, cell proliferation-death regulation, and vascular system development. The mechanisms of these activities were associated with hypoxia-inducible factor-1 (HIF-1), nuclear factor-erythroid 2 related factor 2 (Nrf2), vascular endothelial growth factor (VEGF), forkhead box O (FoxO), phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT), and rhoptry-associated protein1 (Rap1) signaling pathways and their related targets nitric oxide synthase 3 (NOS3), mitogen-activated protein kinase 8 (MAPK8), prostaglandin-endoperoxide synthase 2 (PTGS2), and heme oxygenase 1 (HMOX1). In conclusion, a systematic perspective for the mechanism of TBHQ against DR was revealed by present study which lays a foundation for the application of TBHQ in treating DR.
Collapse
Affiliation(s)
- Chaoqun Liu
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Yue Ren
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xin Sui
- Information and Educational Technology Center, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Kaiyang Liu
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jianing Zhang
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Zian Wang
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Yingying Li
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Yanling Zhang
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
17
|
Giblin MJ, Ontko CD, Penn JS. Effect of cytokine-induced alterations in extracellular matrix composition on diabetic retinopathy-relevant endothelial cell behaviors. Sci Rep 2022; 12:12955. [PMID: 35902594 PMCID: PMC9334268 DOI: 10.1038/s41598-022-12683-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 05/10/2022] [Indexed: 12/15/2022] Open
Abstract
Retinal vascular basement membrane (BM) thickening is an early structural abnormality of diabetic retinopathy (DR). Recent studies suggest that BM thickening contributes to the DR pathological cascade; however, much remains to be elucidated about the exact mechanisms by which BM thickening develops and subsequently drives other pathogenic events in DR. Therefore, we undertook a systematic analysis to understand how human retinal microvascular endothelial cells (hRMEC) and human retinal pericytes (hRP) change their expression of key extracellular matrix (ECM) constituents when treated with diabetes-relevant stimuli designed to model the three major insults of the diabetic environment: hyperglycemia, dyslipidemia, and inflammation. TNFα and IL-1β caused the most potent and consistent changes in ECM expression in both hRMEC and hRP. We also demonstrate that conditioned media from IL-1β-treated human Müller cells caused dose-dependent, significant increases in collagen IV and agrin expression in hRMEC. After narrowing our focus to inflammation-induced changes, we sought to understand how ECM deposited by hRMEC and hRP under inflammatory conditions affects the behavior of naïve hRMEC. Our data demonstrated that diabetes-relevant alterations in ECM composition alone cause both increased adhesion molecule expression by and increased peripheral blood mononuclear cell (PBMC) adhesion to naïve hRMEC. Taken together, these data demonstrate novel roles for inflammation and pericytes in driving BM pathology and suggest that inflammation-induced ECM alterations may advance other pathogenic behaviors in DR, including leukostasis.
Collapse
Affiliation(s)
- Meredith J Giblin
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, USA.
| | - Cayla D Ontko
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, USA
| | - John S Penn
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, USA
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, USA
| |
Collapse
|
18
|
Pan Y, Liu J, Ren J, Luo Y, Sun X. Epac: A Promising Therapeutic Target for Vascular Diseases: A Review. Front Pharmacol 2022; 13:929152. [PMID: 35910387 PMCID: PMC9330031 DOI: 10.3389/fphar.2022.929152] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular diseases affect the circulatory system and comprise most human diseases. They cause severe symptoms and affect the quality of life of patients. Recently, since their identification, exchange proteins directly activated by cAMP (Epac) have attracted increasing scientific interest, because of their role in cyclic adenosine monophosphate (cAMP) signaling, a well-known signal transduction pathway. The role of Epac in cardiovascular disease and cancer is extensively studied, whereas their role in kidney disease has not been comprehensively explored yet. In this study, we aimed to review recent studies on the regulatory effects of Epac on various vascular diseases, such as cardiovascular disease, cerebrovascular disease, and cancer. Accumulating evidence has shown that both Epac1 and Epac2 play important roles in vascular diseases under both physiological and pathological conditions. Additionally, there has been an increasing focus on Epac pharmacological modulators. Therefore, we speculated that Epac could serve as a novel therapeutic target for the treatment of vascular diseases.
Collapse
Affiliation(s)
- Yunfeng Pan
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Jia Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jiahui Ren
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Yun Luo
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Xiaobo Sun
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
19
|
Huang J, Zhou Q. Gene Biomarkers Related to Th17 Cells in Macular Edema of Diabetic Retinopathy: Cutting-Edge Comprehensive Bioinformatics Analysis and In Vivo Validation. Front Immunol 2022; 13:858972. [PMID: 35651615 PMCID: PMC9149582 DOI: 10.3389/fimmu.2022.858972] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Abstract
Background Previous studies have shown that T-helper 17 (Th17) cell-related cytokines are significantly increased in the vitreous of proliferative diabetic retinopathy (PDR), suggesting that Th17 cells play an important role in the inflammatory response of diabetic retinopathy (DR), but its cell infiltration and gene correlation in the retina of DR, especially in diabetic macular edema (DME), have not been studied. Methods The dataset GSE160306 was downloaded from the Gene Expression Omnibus (GEO) database, which contains 9 NPDR samples and 10 DME samples. ImmuCellAI algorithm was used to estimate the abundance of Th17 cells in 24 kinds of infiltrating immune cells. The differentially expressed Th17 related genes (DETh17RGs) between NPDR and DME were documented by difference analysis and correlation analysis. Through aggregate analyses such as gene ontology (GO) and Kyoto Encyclopedia of Gene and Genome (KEGG) pathway enrichment analysis, a protein-protein interaction (PPI) network was constructed to analyze the potential function of DETh17RGs. CytoHubba plug-in algorithm, Lasso regression analysis and support vector machine recursive feature elimination (SVM-RFE) were implemented to comprehensively identify Hub DETh17RGs. The expression archetypes of Hub DETh17RGs were further verified in several other independent datasets related to DR. The Th17RG score was defined as the genetic characterization of six Hub DETh17RGs using the GSVA sample score method, which was used to distinguish early and advanced diabetic nephropathy (DN) as well as normal and diabetic nephropathy. Finally, real-time quantitative PCR (qPCR) was implemented to verify the transcription levels of Hub DETh17RGs in the STZ-induced DR model mice (C57BL/6J). Results 238 DETh17RGs were identified, of which 212 genes were positively correlated while only 26 genes were negatively correlated. Six genes (CD44, CDC42, TIMP1, BMP7, RHOC, FLT1) were identified as Hub DETh17RGs. Because DR and DN have a strong correlation in clinical practice, the verification of multiple independent datasets related to DR and DN proved that Hub DETh17RGs can not only distinguish PDR patients from normal people, but also distinguish DN patients from normal people. It can also identify the initial and advanced stages of the two diseases (NPDR vs DME, Early DN vs Advanced DN). Except for CDC42 and TIMP1, the qPCR transcription levels and trends of other Hub DETh17RGs in STZ-induced DR model mice were consistent with the human transcriptome level in this study. Conclusion This study will improve our understanding of Th17 cell-related molecular mechanisms in the progression of DME. At the same time, it also provides an updated basis for the molecular mechanism of Th17 cell crosstalk in the eye and kidney in diabetes.
Collapse
Affiliation(s)
- Jing Huang
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Jiangxi Center of National Ocular Disease Clinical Research Center, Nanchang, China
| | - Qiong Zhou
- Department of Ophthalmology, The First Affiliated Hospital of Nanchang University, Jiangxi Center of National Ocular Disease Clinical Research Center, Nanchang, China
| |
Collapse
|
20
|
Abdelrahman AA, Powell FL, Jadeja RN, Jones MA, Thounaojam MC, Bartoli M, Al-Shabrawey M, Martin PM. Expression and activation of the ketone body receptor HCAR2/GPR109A promotes preservation of retinal endothelial cell barrier function. Exp Eye Res 2022; 221:109129. [DOI: 10.1016/j.exer.2022.109129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/02/2022] [Accepted: 05/23/2022] [Indexed: 11/30/2022]
|
21
|
Rasmussen M, Zhou J, Schwede F, Ekström P. Enhanced cGMP Interactor Rap Guanine Exchange Factor 4 (EPAC2) Expression and Activity in Degenerating Photoreceptors: A Neuroprotective Response? Int J Mol Sci 2022; 23:ijms23094619. [PMID: 35563009 PMCID: PMC9103912 DOI: 10.3390/ijms23094619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/17/2022] [Accepted: 04/19/2022] [Indexed: 11/25/2022] Open
Abstract
The disease retinitis pigmentosa (RP) leads to photoreceptor degeneration by a yet undefined mechanism(s). In several RP mouse models (i.e., rd mice), a high cyclic GMP (cGMP) level within photoreceptors is detected, suggesting that cGMP plays a role in degeneration. The rap guanine exchange factor 4 (EPAC2) is activated by cyclic AMP (cAMP) and is an accepted cGMP-interacting protein. It is unclear whether and how cGMP interacts with EPAC2 in degenerating photoreceptors; we therefore investigated EPAC2 expression and interactions with cGMP and cAMP in retinas of the rd1 and rd10 models for retinal degeneration. EPAC2 expression in the photoreceptor layer increased significantly during rd1 and rd10 degeneration, and an increase in EPAC2 interactions with cGMP but not cAMP in the rd1 was also seen via a proximity ligation assay on histological sections. Retinal explant cultures revealed that pharmacological inhibition of the EPAC2 activity reduced the photoreceptor layer thickness in the rd10 retina, suggesting that EPAC2 inhibition promotes degeneration. Taken together, our results support the hypothesis that high degeneration-related cGMP leads to increased EPAC2 and cGMP interactions, inhibiting EPAC2. By inference, EPAC2 could have neuroprotective capacities that may be exploited in the future.
Collapse
Affiliation(s)
- Michel Rasmussen
- Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, 22184 Lund, Sweden; (J.Z.); (P.E.)
- Correspondence:
| | - Jiaming Zhou
- Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, 22184 Lund, Sweden; (J.Z.); (P.E.)
| | - Frank Schwede
- BIOLOG Life Science Institute GmbH & Co. KG, 28199 Bremen, Germany;
| | - Per Ekström
- Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, 22184 Lund, Sweden; (J.Z.); (P.E.)
| |
Collapse
|
22
|
Wang C, Li Y, Liu W, Wang C, Jiang W, Zhao Z, Li X, Qiao Y, Dai C. The expression and significance of Epac1 and Epac2 in the inner ear of guinea pigs. Eur Arch Otorhinolaryngol 2022; 279:5207-5214. [PMID: 35438345 DOI: 10.1007/s00405-022-07380-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/28/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To detect the expression of Epac1 and Epac2 in the inner ear of guinea pigs and its association with microcirculation in the inner ear. METHODS The temporal bones of 30 healthy red-eye guinea pigs (60 ears) weighing 200-350 g were collected, then the surrounding bone wall of the cochlea was removed under a dissection microscope. Real-time quantitative PCR (RT-qPCR) and Western blot were used to detect mRNA and protein expression, respectively, of Epac1 and Epac2 in the inner ear and to compare their expression in heart, liver, kidney, intestine, and lung tissues. The specimens of the cochlea included the stria vascularis, basilar membrane, saccule, and utricles isolated under a microscope to detect the localization of Epac1 and Epac2 proteins in various parts of the inner ear through immunofluorescence staining. RESULTS The RT-qPCR and Western blot results showed that Epac1 mRNA was universally expressed in the inner ear, heart, liver, kidneys, intestines, and lungs, and was highly expressed in the liver, kidneys, and intestines (p < 0.05 vs heart, liver, kidney, intestine; p > 0.05 vs lung). Epac2 mRNA was expressed in the inner ear and heart, but not in the liver, kidneys, intestines, or lungs (p < 0.05 vs Heart). Epac1 and Epac2 proteins were both expressed in the inner ear, heart, liver, kidneys, intestines, and lungs. The relative expression of Epac1 proteins in the inner ear was significantly different from the liver, kidneys, intestines, and lungs (p < 0.05). The relative expression of Epac2 proteins in the inner ear was significantly different from the liver, kidneys, and lungs (p < 0.05), but not from the heart (p = 0.127) or intestines (p = 0.274). Immunofluorescence staining observed under confocal microscopy indicated that Epac1 and Epac2 proteins were expressed in the stria vascularis, basilar membrane, saccule, and utricles of the inner ear. They were expressed in maginal cells, intermediate cells, and basal cells of the stria vascularis, and highly expressed in capillary endothelial cells. CONCLUSIONS Epac1 and Epac2 mRNA and proteins were both expressed in the inner ear of guinea pigs and evenly expressed in the spiral ganglion, basilar membrane, saccule, and utricles. However, their expression in capillary endothelial cells of the stria vascularis was more obvious, suggesting that cyclic adenosine monophosphate-Epac1 signaling may play an important role in maintaining the function of the blood-labyrinth barrier and regulating the stability of microcirculation in the inner ear.
Collapse
Affiliation(s)
- Chuan Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, People's Republic of China.,Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, 221000, People's Republic of China.,Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, 221002, People's Republic of China
| | - Yuan Li
- Department of Radiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Wen Liu
- Department of Otorhinolaryngology-Head and Neck Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, People's Republic of China
| | - Caiji Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, People's Republic of China.,Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, 221000, People's Republic of China.,Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, 221002, People's Republic of China
| | - Wen Jiang
- Department of Otorhinolaryngology-Head and Neck Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, People's Republic of China.,Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, 221000, People's Republic of China.,Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, 221002, People's Republic of China
| | - Zeqi Zhao
- Department of Otorhinolaryngology-Head and Neck Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, People's Republic of China.,Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, 221000, People's Republic of China.,Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, 221002, People's Republic of China
| | - Xuanyi Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, People's Republic of China. .,Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, 221000, People's Republic of China. .,Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, 221002, People's Republic of China.
| | - Yuehua Qiao
- Department of Otorhinolaryngology-Head and Neck Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, People's Republic of China.,Institute of Audiology and Balance Science, Xuzhou Medical University, Xuzhou, 221000, People's Republic of China.,Artificial Auditory Laboratory of Jiangsu Province, Xuzhou Medical University, Xuzhou, 221002, People's Republic of China
| | - Chunfu Dai
- National Health Commission Key Laboratory of Hearing Medicine (Fudan University), Department of Otology and Skull Base Surgery, Shanghai, 200000, People's Republic of China
| |
Collapse
|
23
|
Khan A, Ni W, Baltazar T, Lopez-Giraldez F, Pober JS, Pierce RW. ArhGEF12 activates Rap1A and not RhoA in human dermal microvascular endothelial cells to reduce tumor necrosis factor-induced leak. FASEB J 2022; 36:e22254. [PMID: 35294066 PMCID: PMC9103844 DOI: 10.1096/fj.202101873rr] [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: 12/06/2021] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 12/31/2022]
Abstract
Overwhelming inflammation in the setting of acute critical illness induces capillary leak resulting in hypovolemia, edema, tissue dysoxia, organ failure and even death. The tight junction (TJ)-dependent capillary barrier is regulated by small GTPases, but the specific regulatory molecules most active in this vascular segment under such circumstances are not well described. We set out to identify GTPase regulatory molecules specific to endothelial cells (EC) that form TJs. Transcriptional profiling of confluent monolayers of TJ-forming human dermal microvascular ECs (HDMECs) and adherens junction only forming-human umbilical vein EC (HUVECs) demonstrate ARHGEF12 is basally expressed at higher levels and is only downregulated in HDMECs by junction-disrupting tumor necrosis factor (TNF). HDMECs depleted of ArhGEF12 by siRNA demonstrate a significantly exacerbated TNF-induced decrease in trans-endothelial electrical resistance and disruption of TJ continuous staining. ArhGEF12 is established as a RhoA-GEF in HUVECs and its knock down would be expected to reduce RhoA activity and barrier disruption. Pulldown of active GEFs from HDMECs depleted of ArhGEF12 and treated with TNF show decreased GTP-bound Rap1A after four hours but increased GTP-bound RhoA after 12 h. In cell-free assays, ArhGEF12 immunoprecipitated from HDMECs is able to activate both Rap1A and RhoA, but not act on Rap2A-C, RhoB-C, or even Rap1B which shares 95% sequence identity with Rap1A. We conclude that in TJ-forming HDMECs, ArhGEF12 selectively activates Rap1A to limit capillary barrier disruption in a mechanism independent of cAMP-mediated Epac1 activation.
Collapse
Affiliation(s)
- Alamzeb Khan
- Department of Pediatrics, Yale School of Medicine, Yale University
| | - Weiming Ni
- Department of Pediatrics, Yale School of Medicine, Yale University
| | - Tania Baltazar
- Department of Immunobiology, Yale School of Medicine, Yale University
| | | | - Jordan S. Pober
- Department of Immunobiology, Yale School of Medicine, Yale University
| | | |
Collapse
|
24
|
Epigenome-Wide Association Study Reveals Differential Methylation Sites and Association of Gene Expression Regulation with Ischemic Moyamoya Disease in Adults. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7192060. [PMID: 35368875 PMCID: PMC8970806 DOI: 10.1155/2022/7192060] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/28/2022] [Indexed: 12/12/2022]
Abstract
Background The association of DNA methylation with the pathogenesis of adult ischemic moyamoya disease (MMD) is unknown. Here, we investigated the genome-wide DNA methylation profiles in patients with MMD and identified the genes related to the pathogenesis of MMD. Methods Whole blood samples were collected from 20 individuals, including 10 patients with ischemic moyamoya disease without any underlying disease and 10 healthy individuals. Genome-wide DNA methylation analysis was performed using Illumina 850K microarrays. Transcriptional correlation was verified using quantitative reverse transcription-polymerase chain reaction. In vitro experiments were used to analyze the association of functional defects with candidate epigenetic markers. Results The genome-wide methylation level in the whole blood of adults with ischemic MMD was higher than that in the healthy individuals. In total, 759 methylation probes differed significantly between the case and control. The hypermethylated regions were mostly concentrated in the gene spacer regions. Among genes with the highest degree of the differential expression, KCNMA1 and GALNT2 were upregulated, whereas SOX6 and RBM33 were downregulated. Conclusions This is the first study showing that the low expression of genes associated with epigenetic regulation, such as SOX6 and RBM33, may be related to vascular occlusion in MMD, whereas the overexpression of KCNMA1 and GALNT2 may be related to the vascular hyperplasia. The results suggest that DNA methylation was involved in the pathogenesis of MMD, and new pathogenic genes were proposed as biological markers.
Collapse
|
25
|
Huang J, Zhou Q. CD8+T Cell-Related Gene Biomarkers in Macular Edema of Diabetic Retinopathy. Front Endocrinol (Lausanne) 2022; 13:907396. [PMID: 35937822 PMCID: PMC9355330 DOI: 10.3389/fendo.2022.907396] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND CD8+T lymphocytes have a strong pro-inflammatory effect in all parts of the tissue, and some studies have demonstrated that its concentration in the vitreous increased significantly, suggesting that CD8+T cells play a pivotal role in the inflammatory response of diabetic retinopathy (DR). However, the infiltration of CD8+T cells in the DR retina, especially in diabetic macular edema (DME), and its related genes are still unclear. METHODS Download the GSE16036 dataset from the Gene Expression Omnibus (GEO) database. The ImmuCellAI program was performed to evaluate the abundance of 24 immune cells including CD8+T cells. The CD8+T cell-related genes (DECD8+TRGs) between non-proliferative diabetic retinopathy (NPDR) and DME were detected via difference analysis and correlation analysis. Enrichment analysis and protein-protein interaction (PPI) network mapping were implemented to explore the potential function of DECD8+TRGs. Lasso regression, support vector machine recursive feature elimination (SVM-RFE), CytoHubba plug-in and MCODE plug-in in Cytoscape software, and Weighted Gene Co-Expression Network Analysis (WGCNA) were performed to comprehensively analyze and obtain Hub DECD8+TRGs. Hub DECD8+TRGs expression patterns were further validated in other two DR-related independent datasets. The CD8+TRG score was defined as the genetic characterization of Hub DECD8+TRGs using the GSVA sample scoring method, which can be administered to distinguish early and advanced diabetic nephropathy (DN) as well as normal and DN. Finally, the transcription level of DECD8+TRGs in DR model mouse were verified by quantitative real-time PCR (qPCR). RESULTS A total of 371 DECD8+TRGs were identified, of which 294 genes were positively correlated and only 77 genes were negatively correlated. Eight genes (IKZF1, PTPRC, ITGB2, ITGAX, TLR7, LYN, CD74, SPI1) were recognized as Hub DECD8+TRGs. DR and DN, which have strong clinical correlation, have been proved to be associated with CD8+T cell-related hub genes by multiple independent data sets. Hub DECD8+TRGs can not only distinguish PDR from normal and DN from normal, but also play a role in the early and progressive stages of the two diseases (NPDR vs DME, Early DN vs Advanced DN). The qPCR transcription level and trend of Hub DECD8+TRGs in DR mouse model was basically the same as that in human transcriptome. CONCLUSION This study not only increases our understanding of the molecular mechanism of CD8+T cells in the progression of DME, but also expands people's cognitive vision of the molecular mechanism of crosstalk of CD8+T cells in the eyes and kidneys of patients with diabetes.
Collapse
|
26
|
Yamamoto K, Takagi Y, Ando K, Fukuhara S. Rap1 Small GTPase Regulates Vascular Endothelial-Cadherin-Mediated Endothelial Cell-Cell Junctions and Vascular Permeability. Biol Pharm Bull 2021; 44:1371-1379. [PMID: 34602545 DOI: 10.1248/bpb.b21-00504] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The vascular permeability of the endothelium is finely controlled by vascular endothelial (VE)-cadherin-mediated endothelial cell-cell junctions. In the majority of normal adult tissues, endothelial cells in blood vessels maintain vascular permeability at a relatively low level, while in response to inflammation, they limit vascular barrier function to induce plasma leakage and extravasation of immune cells as a defense mechanism. Thus, the dynamic but also simultaneously tight regulation of vascular permeability by endothelial cells is responsible for maintaining homeostasis and, as such, impairments of its underlying mechanisms result in hyperpermeability, leading to the development and progression of various diseases including coronavirus disease 2019 (COVID-19), a newly emerging infectious disease. Recently, increasing numbers of studies have been unveiling the important role of Rap1, a small guanosine 5'-triphosphatase (GTPase) belonging to the Ras superfamily, in the regulation of vascular permeability. Rap1 enhances VE-cadherin-mediated endothelial cell-cell junctions to potentiate vascular barrier functions via dynamic reorganization of the actin cytoskeleton. Importantly, Rap1 signaling activation reportedly improves vascular barrier function in animal models of various diseases associated with vascular hyperpermeability, suggesting that Rap1 might be an ideal target for drugs intended to prevent vascular barrier dysfunction. Here, we describe recent progress in understanding the mechanisms by which Rap1 potentiates VE-cadherin-mediated endothelial cell-cell adhesions and vascular barrier function. We also discuss how alterations in Rap1 signaling are related to vascular barrier dysfunction in diseases such as acute pulmonary injury and malignancies. In addition, we examine the possibility of Rap1 signaling as a target of drugs for treating diseases associated with vascular hyperpermeability.
Collapse
Affiliation(s)
- Kiyotake Yamamoto
- Department of Molecular Pathophysiology, Institute for Advanced Medical Sciences, Nippon Medical School
| | - Yuki Takagi
- Department of Molecular Pathophysiology, Institute for Advanced Medical Sciences, Nippon Medical School
| | - Koji Ando
- Department of Molecular Pathophysiology, Institute for Advanced Medical Sciences, Nippon Medical School
| | - Shigetomo Fukuhara
- Department of Molecular Pathophysiology, Institute for Advanced Medical Sciences, Nippon Medical School
| |
Collapse
|
27
|
Nashine S. Potential Therapeutic Candidates for Age-Related Macular Degeneration (AMD). Cells 2021; 10:cells10092483. [PMID: 34572131 PMCID: PMC8464988 DOI: 10.3390/cells10092483] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 12/27/2022] Open
Abstract
Aging contributes to the risk of development of ocular diseases including, but not limited to, Age-related Macular Degeneration (AMD) that is a leading cause of blindness in the United States as well as worldwide. Retinal aging, that contributes to AMD pathogenesis, is characterized by accumulation of drusen deposits, alteration in the composition of Bruch’s membrane and extracellular matrix, vascular inflammation and dysregulation, mitochondrial dysfunction, and accumulation of reactive oxygen species (ROS), and subsequent retinal pigment epithelium (RPE) cell senescence. Since there are limited options available for the prophylaxis and treatment of AMD, new therapeutic interventions are constantly being looked into to identify new therapeutic targets for AMD. This review article discusses the potential candidates for AMD therapy and their known mechanisms of cytoprotection in AMD. These target therapeutic candidates include APE/REF-1, MRZ-99030, Ciliary NeuroTrophic Factor (CNTF), RAP1 GTPase, Celecoxib, and SS-31/Elamipretide.
Collapse
Affiliation(s)
- Sonali Nashine
- Department of Ophthalmology, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
28
|
Regulation of Rac1 Activation in Choroidal Endothelial Cells: Insights into Mechanisms in Age-Related Macular Degeneration. Cells 2021; 10:cells10092414. [PMID: 34572063 PMCID: PMC8469925 DOI: 10.3390/cells10092414] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/11/2022] Open
Abstract
Age-related macular degeneration (AMD) is one of the leading causes of blindness worldwide. Vision loss from the neovascular form is associated with the invasion of choroidal endothelial cells into the neural retina to form vision-threatening macular neovascularization (MNV). Anti-angiogenic agents are the current standard of care but are effective in only ~50% of AMD cases. The molecular mechanisms involved in invasive MNV point to the importance of regulating signaling pathways that lead to pathologic biologic outcomes. In studies testing the effects of AMD-related stresses, activation of the Rho GTPase, Rac1, was found to be important for the choroidal endothelial cell invasion into the neural retina. However, current approaches to prevent Rac1 activation are inefficient and less effective. We summarize active Rac1-mediated mechanisms that regulate choroidal endothelial cell migration. Specifically, we discuss our work regarding the role of a multidomain protein, IQ motif containing GTPase activating protein 1 (IQGAP1), in sustaining pathologic Rac1 activation and a mechanism by which active Rap1, a Ras-like GTPase, may prevent active Rac1-mediated choroidal endothelial cell migration.
Collapse
|
29
|
Li Y, Baccouche B, Olayinka O, Serikbaeva A, Kazlauskas A. The Role of the Wnt Pathway in VEGF/Anti-VEGF-Dependent Control of the Endothelial Cell Barrier. Invest Ophthalmol Vis Sci 2021; 62:17. [PMID: 34542556 PMCID: PMC8458780 DOI: 10.1167/iovs.62.12.17] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Purpose Investigate the contribution of the Wnt pathway to vascular endothelial growth factor (VEGF)/anti-VEGF-mediated control of endothelial cell permeability. Methods High glucose-treated primary human retinal endothelial cells (HRECs) were exposed to either VEGF, or VEGF and then anti-VEGF. Changes in gene expression were assayed by RNAseq and qRT-PCR. Permeability was monitored by electrical cell-substrate impedance sensing (ECIS). Approaches to activate the Wnt pathway included treatment with LiCl and overexpression of constitutively activated β-catenin. β-catenin-dependent transcriptional activity was monitored in HRECs stably expressing a TCF/LEF-driven reporter. Results VEGF/anti-VEGF altered expression of genes encoding many members of the Wnt pathway. A subset of these genes was regulated in a way that is likely to contribute to control of the endothelial cell barrier. Namely, the VEGF-induced alteration of expression of such genes was reversed by anti-VEGF, and such adjustments occurred at times corresponding to changes in barrier function. While pharmacological and molecular approaches to activate the Wnt pathway had no effect on basal permeability, they suppressed VEGF-induced relaxation. Furthermore, anti-VEGF-mediated restoration of barrier function was unaffected by activation of the Wnt pathway. Conclusions VEGF/anti-VEGF engages multiple members of the Wnt pathway, and activating this pathway enforces the endothelial barrier by attenuating VEGF-induced relaxation. These data suggest that FDA-approved agents such as LiCl may be an adjuvant to anti-VEGF therapy for patients afflicted with blinding conditions including diabetic retinopathy.
Collapse
Affiliation(s)
- Yueru Li
- University of Illinois at Chicago, Department of Ophthalmology & Visual Sciences, Chicago, IL, United States
| | - Basma Baccouche
- University of Illinois at Chicago, Department of Ophthalmology & Visual Sciences, Chicago, IL, United States
| | - Olamide Olayinka
- Department of Physiology and Biophysics, Chicago, IL, United States
| | - Anara Serikbaeva
- Department of Physiology and Biophysics, Chicago, IL, United States
| | - Andrius Kazlauskas
- University of Illinois at Chicago, Department of Ophthalmology & Visual Sciences, Chicago, IL, United States.,Department of Physiology and Biophysics, Chicago, IL, United States
| |
Collapse
|
30
|
Saccharin and Sucralose Protect the Glomerular Microvasculature In Vitro against VEGF-Induced Permeability. Nutrients 2021; 13:nu13082746. [PMID: 34444906 PMCID: PMC8401733 DOI: 10.3390/nu13082746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/02/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
Diabetic kidney disease (DKD) has become a global health concern, with about 40% of people living with type 1 and type 2 diabetes mellitus developing DKD. Upregulation of vascular endothelial growth factor (VEGF) in the kidney is a significant pathology of DKD associated with increased glomerular vascular permeability. To date, however, current anti-VEGF therapies have demonstrated limited success in treating DKD. Recent studies have shown that artificial sweeteners exhibit anti-VEGF potential. The aim of this study was therefore to assess the effects of aspartame, saccharin, and sucralose on VEGF-induced leak using an in vitro model of the glomerular endothelium. Saccharin and sucralose but not aspartame protected against VEGF-induced permeability. Whilst the sweeteners had no effect on traditional VEGF signalling, GC-MS analysis demonstrated that the sweetener sucralose was not able to enter the glomerular endothelial cell to exert the protective effect. Chemical and molecular inhibition studies demonstrated that sweetener-mediated protection of the glomerular endothelium against VEGF is dependent on the sweet taste receptor, T1R3. These studies demonstrate the potential for sweeteners to exert a protective effect against VEGF-induced increased permeability to maintain a healthy endothelium and protect against vascular leak in the glomerulus in settings of DKD.
Collapse
|
31
|
cAMP Compartmentalization in Cerebrovascular Endothelial Cells: New Therapeutic Opportunities in Alzheimer's Disease. Cells 2021; 10:cells10081951. [PMID: 34440720 PMCID: PMC8392343 DOI: 10.3390/cells10081951] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/19/2021] [Accepted: 07/28/2021] [Indexed: 12/20/2022] Open
Abstract
The vascular hypothesis used to explain the pathophysiology of Alzheimer’s disease (AD) suggests that a dysfunction of the cerebral microvasculature could be the beginning of alterations that ultimately leads to neuronal damage, and an abnormal increase of the blood–brain barrier (BBB) permeability plays a prominent role in this process. It is generally accepted that, in physiological conditions, cyclic AMP (cAMP) plays a key role in maintaining BBB permeability by regulating the formation of tight junctions between endothelial cells of the brain microvasculature. It is also known that intracellular cAMP signaling is highly compartmentalized into small nanodomains and localized cAMP changes are sufficient at modifying the permeability of the endothelial barrier. This spatial and temporal distribution is maintained by the enzymes involved in cAMP synthesis and degradation, by the location of its effectors, and by the existence of anchor proteins, as well as by buffers or different cytoplasm viscosities and intracellular structures limiting its diffusion. This review compiles current knowledge on the influence of cAMP compartmentalization on the endothelial barrier and, more specifically, on the BBB, laying the foundation for a new therapeutic approach in the treatment of AD.
Collapse
|
32
|
Liu S, Wang C, Lu J, Dai G, Che H, He W. Long-term inhibition of UCHL1 decreases hypertension and retinopathy in spontaneously hypertensive rats. J Int Med Res 2021; 49:3000605211020641. [PMID: 34130526 PMCID: PMC8212382 DOI: 10.1177/03000605211020641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE To investigate the role of the deubiquitinase ubiquitin C-terminal hydrolase L1 (UCHL1) in hypertension and retinopathy in the spontaneously hypertensive rat (SHR). METHODS Wistar-Kyoto (WKY) rats and SHRs were administered the UCHL1 inhibitor LDN57444 (20 μg/kg/day) for 4 months. Pathological changes were detected with hematoxylin and eosin, immunofluorescence, and dihydroethidium staining. The mRNA and protein expression of UCHL1 were examined by real-time PCR and immunoblotting analysis. RESULTS At 6 months of age, SHRs showed significantly increased mRNA and protein levels of UCHL1 in the retina compared with WKY rats. Moreover, SHRs exhibited significantly increased central retinal thickness, inflammation, and reactive oxygen species production compared with WKY rats, and these effects were markedly attenuated by systemic administration of the UCHL1 inhibitor LDN57444. The beneficial effects of LDN57444 were possibly associated with reduced blood pressure and the inactivation of several signaling pathways. CONCLUSION UCHL1 is involved in hypertension and retinopathy in SHRs, suggesting that UCHL1 may be used as a potential therapeutic target for treating hypertensive retinopathy.
Collapse
Affiliation(s)
- Shasha Liu
- The Second Clinical College, Dalian Medical University, Dalian, P.R. China.,Health Management Center, First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| | - Chengfang Wang
- Health Management Center, First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| | - Jianmin Lu
- Department of Ophthalmology, First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| | - Guangzheng Dai
- Clinical Research Center, He Eye Specialists Hospitals, Shenyang, P.R. China
| | - Huixin Che
- Clinical Research Center, He Eye Specialists Hospitals, Shenyang, P.R. China
| | - Wei He
- The Second Clinical College, Dalian Medical University, Dalian, P.R. China.,Clinical Research Center, He Eye Specialists Hospitals, Shenyang, P.R. China
| |
Collapse
|
33
|
Chu W, Sun X, Zhu X, Zhao YC, Zhang J, Kong Q, Zhou L. Blockade of platelet glycoprotein receptor Ib ameliorates blood-brain barrier disruption following ischemic stroke via Epac pathway. Biomed Pharmacother 2021; 140:111698. [PMID: 34029954 DOI: 10.1016/j.biopha.2021.111698] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 04/04/2021] [Accepted: 04/21/2021] [Indexed: 10/21/2022] Open
Abstract
Glycoprotein (GP) Ib is a platelet membrane receptor complex exposed to vascular injury, proposed as an effective target for stroke therapy. Previously, we have observed that the GPIb antagonist anfibatide (ANF) could mitigate blood-brain barrier (BBB) disruption following cerebral ischemia/reperfusion (CI/R) injury. The current study was designed to investigate whether the amelioration of the BBB by ANF is mediated via the Epac signaling pathway. A murine model of CI/R injury was induced following 90 min of transient middle cerebral artery occlusion (MCAO). ANF (4 μg/kg) was intravenously injected 1 h after reperfusion. Herein, ANF ameliorated BBB disruption, increased the expression of tight junction proteins, suppressed F-actin cytoskeleton rearrangement, decreased the permeability of the ischemic brain tissue, and relieved brain edema. ANF-treated mice had smaller infarct volumes and less severe neurological deficits than the MCAO mice. Moreover, the effects of ANF and Epac1 agonists were very similar in the MCAO mice. Epac activation with a cAMP analog, 8-CPT-2'-O-Me-cAMP, mitigated the breakdown of BBB function and CI/R injury. The Epac specific antagonist, ESI-09, worsened barrier damage and cerebral impairment, antagonizing the protective effects afforded by ANF. In addition, ANF upregulated the expression of Epac1 protein in the ischemic cerebral cortex. Collectively, our results indicate that the protective effect of ANF on the BBB after CI/R could be attributed to the activation of the Epac pathway.
Collapse
Affiliation(s)
- Wei Chu
- Department of Pharmacology, School of Basic Medical Science, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei 230032, PR China; Department of Medical College, Shenzhen Polytechnic, Shenzhen 518055, PR China
| | - Xuemei Sun
- Department of Pharmacology, School of Basic Medical Science, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei 230032, PR China; Department of Medical College, Shenzhen Polytechnic, Shenzhen 518055, PR China
| | - Xiaoxiao Zhu
- Department of Pharmacology, School of Basic Medical Science, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei 230032, PR China; Department of Medical College, Shenzhen Polytechnic, Shenzhen 518055, PR China
| | - Yu Chen Zhao
- Department of Mathematics, University of California, Los Angeles, CA 90095, USA
| | - Jingcheng Zhang
- Department of Pharmacology, School of Basic Medical Science, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei 230032, PR China
| | - Qin Kong
- Department of Pharmacology, School of Basic Medical Science, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei 230032, PR China
| | - Lanlan Zhou
- Department of Pharmacology, School of Basic Medical Science, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei 230032, PR China; Department of Medical College, Shenzhen Polytechnic, Shenzhen 518055, PR China.
| |
Collapse
|
34
|
Yu B, Xiao M, Yang F, Xiao J, Zhang H, Su L, Zhang X, Li X. MicroRNA-431-5p encapsulated in serum extracellular vesicles as a biomarker for proliferative diabetic retinopathy. Int J Biochem Cell Biol 2021; 135:105975. [PMID: 33838342 DOI: 10.1016/j.biocel.2021.105975] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/28/2021] [Accepted: 04/03/2021] [Indexed: 01/04/2023]
Abstract
Early diagnosis and precise monitoring of the development of proliferative diabetic retinopathy (PDR) can significantly improve therapeutic strategies and help decrease blindness caused by it. Extracellular vesicles (EVs) were recently found to be involved in intercellular communications and are a potential source for the discovery of novel biomarkers. The current study aims to investigate the effectiveness of microRNAs (miRNAs) encapsulated in small EVs (sEVs) as minimally invasive biomarkers for PDR. SEVs were extracted from plasma of healthy subjects, diabetic patients, nonPDR patients and PDR patients. Then, we performed microarray analysis to determine the miRNA expression profile. MiR-431-5p expression doubled in the PDR patients compared with the healthy controls and the diabetic patients. We further found that miR-431-5p expression was 2.3 times higher in 4-hydroxynonenal treated human retinal capillary endothelial cells (HRCECs) than the control. After transfection with miR-431-5p mimics, proliferation of HRCECs was promoted, while transfection with miR-431-5p inhibitor demonstrated the opposite effect. The present findings indicate that circulating sEVs showed a differential miRNA profile in PDR patients. MiR-431-5p was involved in the pathogenesis of PDR development and may function as a novel biomarker for PDR.
Collapse
Affiliation(s)
- Bo Yu
- Tianjin International Joint Research and Development Center of Ophthalmology and Vision Science, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China
| | - Mengran Xiao
- The Second Hospital of Tianjin Medical University, 23 Pingjiang Road, Hexi District, Tianjin, 300211, China
| | - Fuhua Yang
- Tianjin International Joint Research and Development Center of Ophthalmology and Vision Science, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China
| | - Jing Xiao
- Tianjin International Joint Research and Development Center of Ophthalmology and Vision Science, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China
| | - Hui Zhang
- Tianjin International Joint Research and Development Center of Ophthalmology and Vision Science, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China
| | - Lin Su
- Tianjin International Joint Research and Development Center of Ophthalmology and Vision Science, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China
| | - Xiaomin Zhang
- Tianjin International Joint Research and Development Center of Ophthalmology and Vision Science, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China.
| | - Xiaorong Li
- Tianjin International Joint Research and Development Center of Ophthalmology and Vision Science, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China.
| |
Collapse
|
35
|
Cai R, Dong Y, Fang M, Fan Y, Cheng Z, Zhou Y, Gao J, Han F, Guo C, Ma X. Genome-Wide Association Identifies Risk Pathways for SAPHO Syndrome. Front Cell Dev Biol 2021; 9:643644. [PMID: 33816493 PMCID: PMC8012550 DOI: 10.3389/fcell.2021.643644] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/18/2021] [Indexed: 11/13/2022] Open
Abstract
SAPHO syndrome is a rare chronic inflammatory disease which is characterized by the comprehensive manifestations of bone, joint, and skin. However, little is known about the pathogenesis of SAPHO syndrome. A genome-wide association study (GWAS) of 49 patients and 121 control subjects have primarily focused on identification of common genetic variants associated with SAPHO, the data were analyzed by classical multiple logistic regression. Later, GWAS findings were further validated using whole exome sequencing (WES) in 16 patients and 15 controls to identify potentially functional pathways involved in SAPHO pathogenesis. In general, 40588 SNPs in genomic regions were associated with P < 0.05 after filter process, only 9 SNPs meet the expected cut-off P-value, however, none of them had association with SAPHO syndrome based on published literatures. And then, 15 pathways were found involved in SAPHO pathogenesis, of them, 6 pathways including osteoclast differentiation, bacterial invasion of epithelial cells, et al., had strong association with skin, osteoarticular manifestations of SAPHO or inflammatory reaction based published research. This study identified aberrant osteoclast differentiation and other pathways were involved in SAPHO syndrome. This finding may give insight into the understanding of pathogenic genes of SAPHO and provide the basis for SAPHO research and treatment.
Collapse
Affiliation(s)
- Ruikun Cai
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| | - Yichao Dong
- National Research Institute for Family Planning, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Mingxia Fang
- National Research Institute for Family Planning, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Yuxuan Fan
- National Research Institute for Family Planning, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Zian Cheng
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| | - Yue Zhou
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| | - Jianen Gao
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| | - Feifei Han
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Changlong Guo
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| | - Xu Ma
- National Research Institute for Family Planning, Beijing, China
- National Human Genetic Resources Center, Beijing, China
| |
Collapse
|
36
|
Zhao L, He K, Xiao Q, Liu Q, Luo W, Luo J, Fu H, Li J, Wu X, Du J, Gong Q, Wang X, Yang S. Comparative transcriptome profiles of large and small bodied large-scale loaches cultivated in paddy fields. Sci Rep 2021; 11:4936. [PMID: 33654201 PMCID: PMC7925675 DOI: 10.1038/s41598-021-84519-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 02/03/2021] [Indexed: 01/31/2023] Open
Abstract
Fish culture in paddy fields is a traditional aquaculture mode, which has a long history in East Asia. Large-scale loach (Paramisgurnus dabryanus) fast growth is suitable for paddy fields aquaculture in China. The objective of this study was to identify differential expression genes (DEGs) in the brain, liver and muscle tissues between large (LG, top 5% of maximum total length) and small (SG, top 5% of minimum total length) groups using RNA-seq. In total, 150 fish were collected each week and 450 fish were collected at twelfth week from three paddy fields for all the experimental. Histological observation found that the muscle fibre diameter of LG loaches was greater than that of SG loaches. Transcriptome results revealed that the high expression genes (HEGs) in LG loaches (fold change ≥ 2, p < 0.05) were mainly concentrated in metabolic pathways, such as "Thyroid hormone signalling pathway", "Citrate cycle (TCA cycle)", "Carbon metabolism", "Fatty acid metabolism", and "Cholesterol metabolism", and the HEGs in SG loaches were enriched in the pathways related to environmental information processing such as "Cell adhesion molecules (CAMs)", "ECM- receptor interaction" and "Rap1 signalling pathway"; cellular processes such as "Tight junction", "Focal adhesion", "Phagosome" and "Adherens junction". Furthermore, IGFs gene family may play an important role in loach growth for their different expression pattern between the two groups. These findings can enhance our understanding about the molecular mechanism of different growth and development levels of loaches in paddy fields.
Collapse
Affiliation(s)
- Liulan Zhao
- grid.80510.3c0000 0001 0185 3134College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
| | - Kuo He
- grid.80510.3c0000 0001 0185 3134College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
| | - Qing Xiao
- grid.80510.3c0000 0001 0185 3134College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
| | - Qiao Liu
- grid.80510.3c0000 0001 0185 3134College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
| | - Wei Luo
- grid.80510.3c0000 0001 0185 3134College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
| | - Jie Luo
- grid.80510.3c0000 0001 0185 3134College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
| | - Hongmei Fu
- grid.80510.3c0000 0001 0185 3134College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
| | - Jiayao Li
- grid.412514.70000 0000 9833 2433Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, 200090 China
| | - Xugan Wu
- grid.412514.70000 0000 9833 2433Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, 200090 China
| | - Jun Du
- grid.465230.60000 0004 1777 7721Fisheries Institute, Sichuan Academy of Agricultural Science, Chengdu, 611731 China
| | - Quan Gong
- grid.465230.60000 0004 1777 7721Fisheries Institute, Sichuan Academy of Agricultural Science, Chengdu, 611731 China
| | - Xun Wang
- grid.80510.3c0000 0001 0185 3134College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
| | - Song Yang
- grid.80510.3c0000 0001 0185 3134College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130 China
| |
Collapse
|
37
|
Zeitelhofer M, Adzemovic MZ, Moessinger C, Stefanitsch C, Strell C, Muhl L, Brundin L, Fredriksson L, Olsson T, Eriksson U, Nilsson I. Blocking PDGF-CC signaling ameliorates multiple sclerosis-like neuroinflammation by inhibiting disruption of the blood-brain barrier. Sci Rep 2020; 10:22383. [PMID: 33361796 PMCID: PMC7759579 DOI: 10.1038/s41598-020-79598-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
Disruption of blood–brain barrier (BBB) integrity is a feature of various neurological disorders. Here we found that the BBB is differently affected during the preclinical, progression and remission phase of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). We have identified an upregulation of pro-inflammatory and pro-angiogenic factors in the BBB transcriptome and down-regulation of endothelial tight junction members coinciding with elevated BBB leakage specifically during the progression phase. These changes were antagonized by blocking PDGFRα signaling with the small tyrosine kinase inhibitor imatinib. Moreover, targeting the PDGFRα ligand PDGF-CC using a neutralizing antibody, facilitated recovery of BBB integrity and improvement of EAE symptoms. Intracerebroventricular injection of PDGF-CC induced upregulation, whereas blocking PDGF-CC during EAE led to downregulation of Tnfa and Il1a at the BBB. Our findings suggest that blocking PDGF-CC counteracts fundamental aspects of endothelial cell activation and disruption of the BBB by decreasing Tnfa and Il1a expression. We also demonstrate that both PDGF-CC and its receptor PDGFRα were upregulated in MS lesions indicating that blocking PDGF-CC may be considered a novel treatment for MS.
Collapse
Affiliation(s)
- Manuel Zeitelhofer
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden.
| | - Milena Z Adzemovic
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden.,Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Christine Moessinger
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Christina Stefanitsch
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Carina Strell
- Department of Immunology, Genetics and Pathology, Uppsala University, 75185, Uppsala, Sweden
| | - Lars Muhl
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Lou Brundin
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Linda Fredriksson
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Tomas Olsson
- Neuroimmunology Unit, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Ulf Eriksson
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Ingrid Nilsson
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden.
| |
Collapse
|
38
|
Xiong S, Hong Z, Huang LS, Tsukasaki Y, Nepal S, Di A, Zhong M, Wu W, Ye Z, Gao X, Rao GN, Mehta D, Rehman J, Malik AB. IL-1β suppression of VE-cadherin transcription underlies sepsis-induced inflammatory lung injury. J Clin Invest 2020; 130:3684-3698. [PMID: 32298238 PMCID: PMC7324198 DOI: 10.1172/jci136908] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/09/2020] [Indexed: 12/18/2022] Open
Abstract
Unchecked inflammation is a hallmark of inflammatory tissue injury in diseases such as acute respiratory distress syndrome (ARDS). Yet the mechanisms of inflammatory lung injury remain largely unknown. Here we showed that bacterial endotoxin lipopolysaccharide (LPS) and cecal ligation and puncture-induced (CLP-induced) polymicrobial sepsis decreased the expression of transcription factor cAMP response element binding (CREB) in lung endothelial cells. We demonstrated that endothelial CREB was crucial for VE-cadherin transcription and the formation of the normal restrictive endothelial adherens junctions. The inflammatory cytokine IL-1β reduced cAMP generation and CREB-mediated transcription of VE-cadherin. Furthermore, endothelial cell-specific deletion of CREB induced lung vascular injury whereas ectopic expression of CREB in the endothelium prevented the injury. We also observed that rolipram, which inhibits type 4 cyclic nucleotide phosphodiesterase-mediated (PDE4-mediated) hydrolysis of cAMP, prevented endotoxemia-induced lung vascular injury since it preserved CREB-mediated VE-cadherin expression. These data demonstrate the fundamental role of the endothelial cAMP-CREB axis in promoting lung vascular integrity and suppressing inflammatory injury. Therefore, strategies aimed at enhancing endothelial CREB-mediated VE-cadherin transcription are potentially useful in preventing sepsis-induced lung vascular injury in ARDS.
Collapse
Affiliation(s)
- Shiqin Xiong
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Zhigang Hong
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Long Shuang Huang
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Yoshikazu Tsukasaki
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Saroj Nepal
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Anke Di
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Ming Zhong
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Wei Wu
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Zhiming Ye
- Department of Nephrology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaopei Gao
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Gadiparthi N. Rao
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Dolly Mehta
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Jalees Rehman
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Asrar B. Malik
- Department of Pharmacology and Regenerative Medicine and
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| |
Collapse
|
39
|
Thomas G, Frederick E, Hausburg M, Goldberg L, Hoke M, Roshon M, Mains C, Bar-Or D. The novel immunomodulatory biologic LMWF5A for pharmacological attenuation of the "cytokine storm" in COVID-19 patients: a hypothesis. Patient Saf Surg 2020; 14:21. [PMID: 32431755 PMCID: PMC7220573 DOI: 10.1186/s13037-020-00248-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 05/06/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND A common complication of viral pulmonary infections, such as in the ongoing COVID-19 pandemic, is a phenomenon described as a "cytokine storm". While poorly defined, this hyperinflammatory response results in diffuse alveolar damage. The low molecular weight fraction of commercial human serum albumin (LMWF5A), a novel biologic in development for osteoarthritis, demonstrates beneficial in vitro immunomodulatory effects complimentary to addressing inflammation, thus, we hypothesize that LMWF5A could improve the clinical outcomes of COVID-19 by attenuating hyperinflammation and the potential development of a cytokine storm. PRESENTATION OF THE HYPOTHESIS A variety of human in vitro immune models indicate that LMWF5A reduces the production of pro-inflammatory cytokines implicated in cytokine storm associated with COVID-19. Furthermore, evidence suggests LMWF5A also promotes the production of mediators required for resolving inflammation and enhances the barrier function of endothelial cultures. TESTING THE HYPOTHESIS A randomized controlled trial, to evaluate the safety and efficacy of nebulized LMWF5A in adults with Acute Respiratory Distress Syndrome (ARDS) secondary to COVID-19 infection, was developed and is currently under review by the Food and Drug Administration. IMPLICATIONS OF HYPOTHESIS If successful, this therapy may attenuate the cytokine storm observed in these patients and potentially reduce mortality, increase ventilation free days, improve oxygenation parameters and consequently lessen the burden on patients and the intensive care unit. CONCLUSIONS In conclusion, in vitro findings suggest that the immunomodulatory effects of LMWF5A make it a viable candidate for treating cytokine storm and restoring homeostasis to the immune response in COVID-19.
Collapse
Affiliation(s)
- Gregory Thomas
- Ampio Pharmaceuticals, Inc, 373 Inverness Pkwy #200, Englewood, CO 80112 USA
| | - Elizabeth Frederick
- Ampio Pharmaceuticals, Inc, 373 Inverness Pkwy #200, Englewood, CO 80112 USA
| | - Melissa Hausburg
- Trauma Research Department, Swedish Medical Center, 501 E. Hampden, Englewood, CO 80113 USA
- Trauma Research Department, St. Anthony Hospital, 11600 W 2nd Pl, Lakewood, CO 80228 USA
- Trauma Research Department, Medical City Plano, 3901 W 15th St, Plano, TX 75075 USA
- Trauma Research Department, Penrose Hospital, 2222 N Nevada Ave, Colorado Springs, CO 80907 USA
- Trauma Research Department, Research Medical Center, 2316 E Meyer Blvd, Kansas City, MO 64132 USA
- Trauma Research Department, Wesley Medical Center, 550 N Hillside St, Wichita, KS 67214 USA
| | - Laura Goldberg
- Ampio Pharmaceuticals, Inc, 373 Inverness Pkwy #200, Englewood, CO 80112 USA
| | - Marshall Hoke
- Ampio Pharmaceuticals, Inc, 373 Inverness Pkwy #200, Englewood, CO 80112 USA
| | - Michael Roshon
- Emergency Department, Penrose Hospital, Colorado Springs, Colorado USA
| | | | - David Bar-Or
- Trauma Research Department, Swedish Medical Center, 501 E. Hampden, Englewood, CO 80113 USA
- Trauma Research Department, St. Anthony Hospital, 11600 W 2nd Pl, Lakewood, CO 80228 USA
- Trauma Research Department, Medical City Plano, 3901 W 15th St, Plano, TX 75075 USA
- Trauma Research Department, Penrose Hospital, 2222 N Nevada Ave, Colorado Springs, CO 80907 USA
- Trauma Research Department, Research Medical Center, 2316 E Meyer Blvd, Kansas City, MO 64132 USA
- Trauma Research Department, Wesley Medical Center, 550 N Hillside St, Wichita, KS 67214 USA
- Department of Molecular Biology, Rocky Vista University, 8401 S Chambers Rd, Parker, CO 80134 USA
- Swedish Medical Center, 501 E. Hampden Ave. Rm 4-454, Englewood, CO 80013 USA
| |
Collapse
|
40
|
Steinle JJ. Review: Role of cAMP signaling in diabetic retinopathy. Mol Vis 2020; 26:355-358. [PMID: 32476815 PMCID: PMC7245604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/07/2020] [Indexed: 11/25/2022] Open
Abstract
Despite decades of research, diabetic retinopathy remains the leading cause of blindness in working age adults. Treatments for early phases for the disease remain elusive. One pathway that appears to regulate neuronal, vascular, and inflammatory components of diabetic retinopathy is the cyclic adenosine 3', 5'-monophosphate (cAMP) pathway. In this review, we discuss the current literature on cAMP actions on the retina, with a focus on neurovascular changes commonly associated with preproliferative diabetic retinopathy models.
Collapse
|
41
|
Sivertsen Åsrud K, Bjørnstad R, Kopperud R, Pedersen L, Hoeven B, Karlsen TV, Brekke Rygh C, Curry F, Bakke M, Reed RK, Tenstad O, Døskeland SO. Epac1 null mice have nephrogenic diabetes insipidus with deficient corticopapillary osmotic gradient and weaker collecting duct tight junctions. Acta Physiol (Oxf) 2020; 229:e13442. [PMID: 31943825 DOI: 10.1111/apha.13442] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 01/03/2023]
Abstract
AIM The cAMP-mediator Epac1 (RapGef3) has high renal expression. Preliminary observations revealed increased diuresis in Epac1-/- mice. We hypothesized that Epac1 could restrict diuresis by promoting transcellular collecting duct (CD) water and urea transport or by stabilizing CD paracellular junctions to reduce osmolyte loss from the renal papillary interstitium. METHODS In Epac1-/- and Wt C57BL/6J mice, renal papillae, dissected from snap-frozen kidneys, were assayed for the content of key osmolytes. Cell junctions were analysed by transmission electron microscopy. Urea transport integrity was evaluated by urea loading with 40% protein diet, endogenous vasopressin production was manipulated by intragastric water loading and moderate dehydration and vasopressin type 2 receptors were stimulated selectively by i.p.-injected desmopressin (dDAVP). Glomerular filtration rate (GFR) was estimated as [14 C]inulin clearance. The glomerular filtration barrier was evaluated by urinary albumin excretion and microvascular leakage by the renal content of time-spaced intravenously injected 125 I- and 131 I-labelled albumin. RESULTS Epac1-/- mice had increased diuresis and increased free water clearance under antidiuretic conditions. They had shorter and less dense CD tight junction (TJs) and attenuated corticomedullary osmotic gradient. Epac1-/- mice had no increased protein diet-induced urea-dependent osmotic diuresis, and expressed Wt levels of aquaporin-2 (AQP-2) and urea transporter A1/3 (UT-A1/3). Epac1-/- mice had no urinary albumin leakage and unaltered renal microvascular albumin extravasation. Their GFR was moderately increased, unless when treated with furosemide. CONCLUSION Our results conform to the hypothesis that Epac1-dependent mechanisms protect against diabetes insipidus by maintaining renal papillary osmolarity and the integrity of CD TJs.
Collapse
Affiliation(s)
| | - Ronja Bjørnstad
- Department of Biomedicine Faculty of Medicine University of Bergen Bergen Norway
| | - Reidun Kopperud
- Department of Biomedicine Faculty of Medicine University of Bergen Bergen Norway
| | - Line Pedersen
- Department of Biomedicine Faculty of Medicine University of Bergen Bergen Norway
| | - Barbara Hoeven
- Department of Biomedicine Faculty of Medicine University of Bergen Bergen Norway
| | - Tine V. Karlsen
- Department of Biomedicine Faculty of Medicine University of Bergen Bergen Norway
| | - Cecilie Brekke Rygh
- Department of Biomedicine Faculty of Medicine University of Bergen Bergen Norway
- Faculty of Health and Social Sciences Western Norway University of Applied Sciences Bergen Norway
| | - Fitz‐Roy Curry
- Department of Physiology and Membrane Biology School of Medicine University of California Davis CA USA
| | - Marit Bakke
- Department of Biomedicine Faculty of Medicine University of Bergen Bergen Norway
| | - Rolf K. Reed
- Department of Biomedicine Faculty of Medicine University of Bergen Bergen Norway
- Centre for Cancer Biomarkers University of Bergen Bergen Norway
| | - Olav Tenstad
- Department of Biomedicine Faculty of Medicine University of Bergen Bergen Norway
| | - Stein O. Døskeland
- Department of Biomedicine Faculty of Medicine University of Bergen Bergen Norway
| |
Collapse
|
42
|
Liu X, Wilson MW, Liu K, Lee P, Yeomans L, Hagen SE, Lin CM, Wen B, Sun D, White AD, Showalter HD, Antonetti DA. Synthesis and structure-activity relationships of thieno[2,3-d]pyrimidines as atypical protein kinase C inhibitors to control retinal vascular permeability and cytokine-induced edema. Bioorg Med Chem 2020; 28:115480. [PMID: 32327351 DOI: 10.1016/j.bmc.2020.115480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 11/28/2022]
Abstract
Studies demonstrate that small molecule targeting of atypical protein kinase C (aPKC) may provide an effective means to control vascular permeability, prevent edema, and reduce inflammation providing novel and important alternatives to anti-VEGF therapies for certain blinding eye diseases. Based on a literature tricyclic thieno[2,3-d]pyrimidine lead (1), an ATP-competitive inhibitor of the aPKC iota (ι) and aPKC zeta (ζ) isoforms, we have synthesized a small series of compounds in 1-2 steps from a readily available chloro intermediate. A single pyridine congener was also made using 2D NMR to assign regiochemistry. Within the parent pyrimidine series, a range of potencies was observed against aPKCζ whereas the pyridine congener was inactive. Selected compounds were also tested for their effect toward VEGF-induced permeability in BREC cells. The most potent of these (7l) was further assayed against the aPKCι isoform and showed a favorable selectivity profile against a panel of 31 kinases, including kinases from the AGC superfamily, with a focus on PKC isoforms and kinases previously shown to affect permeability. Further testing of 7l in a luciferase assay in HEK293 cells showed an ability to prevent TNF-α induced NFκB activation while not having any effect on cell survival. Intravitreal administration of 7l to the eye yielded a complete reduction in permeability in a test to determine whether the compound could block VEGF- and TNFα-induced permeability across the retinal vasculature in a rat model. The compound in mice displayed good microsomal stability and in plasma moderate exposure (AUC and Cmax), low clearance, a long half-life and high oral bioavailability. With IV dosing, higher levels were observed in the brain and eye relative to plasma, with highest levels in the eye by either IV or PO dosing. With a slow oral absorption profile, 7l accumulates in the eye to maintain a high concentration after dosing with higher levels than in plasma. Compound 7l may represent a class of aPKC inhibitors for further investigation.
Collapse
Affiliation(s)
- Xuwen Liu
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Michael W Wilson
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA; Vahlteich Medicinal Chemistry Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kun Liu
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA; Vahlteich Medicinal Chemistry Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pil Lee
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA; Vahlteich Medicinal Chemistry Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Larisa Yeomans
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Susan E Hagen
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA; Vahlteich Medicinal Chemistry Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Cheng-Mao Lin
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Bo Wen
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Duxin Sun
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Andrew D White
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA; Vahlteich Medicinal Chemistry Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hollis D Showalter
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - David A Antonetti
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
43
|
Flemming S, Luissint AC, Kusters DHM, Raya-Sandino A, Fan S, Zhou DW, Hasegawa M, Garcia-Hernandez V, García AJ, Parkos CA, Nusrat A. Desmocollin-2 promotes intestinal mucosal repair by controlling integrin-dependent cell adhesion and migration. Mol Biol Cell 2020; 31:407-418. [PMID: 31967937 PMCID: PMC7185897 DOI: 10.1091/mbc.e19-12-0692] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The intestinal mucosa is lined by a single layer of epithelial cells that forms a tight barrier, separating luminal antigens and microbes from underlying tissue compartments. Mucosal damage results in a compromised epithelial barrier that can lead to excessive immune responses as observed in inflammatory bowel disease. Efficient wound repair is critical to reestablish the mucosal barrier and homeostasis. Intestinal epithelial cells (IEC) exclusively express the desmosomal cadherins, Desmoglein-2 and Desmocollin-2 (Dsc2) that contribute to mucosal homeostasis by strengthening intercellular adhesion between cells. Despite this important property, specific contributions of desmosomal cadherins to intestinal mucosal repair after injury remain poorly investigated in vivo. Here we show that mice with inducible conditional knockdown (KD) of Dsc2 in IEC (Villin-CreERT2; Dsc2 fl/fl) exhibited impaired mucosal repair after biopsy-induced colonic wounding and recovery from dextran sulfate sodium-induced colitis. In vitro analyses using human intestinal cell lines after KD of Dsc2 revealed delayed epithelial cell migration and repair after scratch-wound healing assay that was associated with reduced cell–matrix traction forces, decreased levels of integrin β1 and β4, and altered activity of the small GTPase Rap1. Taken together, these results demonstrate that epithelial Dsc2 is a key contributor to intestinal mucosal wound healing in vivo.
Collapse
Affiliation(s)
- Sven Flemming
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | | | | | | | - Shuling Fan
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Dennis W Zhou
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332
| | - Mizuho Hasegawa
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | | | - Andrés J García
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332.,Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332
| | - Charles A Parkos
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Asma Nusrat
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
44
|
Cerebral Cavernous Malformation Proteins in Barrier Maintenance and Regulation. Int J Mol Sci 2020; 21:ijms21020675. [PMID: 31968585 PMCID: PMC7013531 DOI: 10.3390/ijms21020675] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 12/18/2022] Open
Abstract
Cerebral cavernous malformation (CCM) is a disease characterized by mulberry shaped clusters of dilated microvessels, primarily in the central nervous system. Such lesions can cause seizures, headaches, and stroke from brain bleeding. Loss-of-function germline and somatic mutations of a group of genes, called CCM genes, have been attributed to disease pathogenesis. In this review, we discuss the impact of CCM gene encoded proteins on cellular signaling, barrier function of endothelium and epithelium, and their contribution to CCM and potentially other diseases.
Collapse
|
45
|
Liu H, Mei FC, Yang W, Wang H, Wong E, Cai J, Toth E, Luo P, Li YM, Zhang W, Cheng X. Epac1 inhibition ameliorates pathological angiogenesis through coordinated activation of Notch and suppression of VEGF signaling. SCIENCE ADVANCES 2020; 6:eaay3566. [PMID: 31911948 PMCID: PMC6938696 DOI: 10.1126/sciadv.aay3566] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/29/2019] [Indexed: 05/26/2023]
Abstract
In this study, we investigated the roles of Epac1 in pathological angiogenesis and its potential as a novel therapeutic target for the treatment of vasoproliferative diseases. Genetic deletion of Epac1 ameliorated pathological angiogenesis in mouse models of oxygen-induced retinopathy (OIR) and carotid artery ligation. Moreover, genetic deletion or pharmacological inhibition of Epac1 suppressed microvessel sprouting from ex vivo aortic ring explants. Mechanistic studies revealed that Epac1 acted as a previously unidentified inhibitor of the γ-secretase/Notch signaling pathway via interacting with γ-secretase and regulating its intracellular trafficking while enhancing vascular endothelial growth factor signaling to promote pathological angiogenesis. Pharmacological administration of an Epac-specific inhibitor suppressed OIR-induced neovascularization in wild-type mice, recapitulating the phenotype of genetic Epac1 knockout. Our results demonstrate that Epac1 signaling is critical for the progression of pathological angiogenesis but not for physiological angiogenesis and that the newly developed Epac-specific inhibitors are effective in combating proliferative retinopathy.
Collapse
Affiliation(s)
- Hua Liu
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Fang C. Mei
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, TX, USA
| | - Wenli Yang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, TX, USA
| | - Hui Wang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, TX, USA
| | - Eitan Wong
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jingjing Cai
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Emma Toth
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, TX, USA
| | - Pei Luo
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, TX, USA
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wenbo Zhang
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, TX, USA
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
- Texas Therapeutics Institute, University of Texas Health Science Center, Houston, TX, USA
| |
Collapse
|
46
|
Genome-Wide Mapping Defines a Role for C/EBPβ and c-Jun in Non-Canonical Cyclic AMP Signalling. Cells 2019; 8:cells8101253. [PMID: 31615122 PMCID: PMC6829624 DOI: 10.3390/cells8101253] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/11/2019] [Accepted: 10/12/2019] [Indexed: 01/06/2023] Open
Abstract
The novel exchange protein activated by cyclic AMP (EPAC1) activator, I942, induces expression of the suppressor of cytokine signalling 3 (SOCS3) gene, thereby inhibiting interleukin 6 (IL6) inflammatory processes in human umbilical vein endothelial cells (HUVECs). Here we use RNA-SEQ and ChIP-SEQ to determine global gene responses to I942, in comparison with cyclic AMP production promoted by forskolin and rolipram (F/R). We found that I942 promoted significant changes in the RNA expression of 1413 genes, largely associated with microtubule stability and cell cycle progression, whereas F/R regulated 197 genes linked to endothelial cell function, including chemokine production and platelet aggregation. A further 108 genes were regulated by both treatments, including endothelial regulatory genes involved in purinergic signalling and cell junction organization. ChIP-SEQ demonstrated that F/R induced genome-wide recruitment of C/EBPβ and c-Jun transcription factors, whereas I942 promoted recruitment of c-Jun to genes associated with IL6 signalling, with little effect on C/EBPβ activation. Despite this, certain key inflammatory genes, including IL6, VEGF, CCL2/MCP1, VCAM1, SELE and ICAM1 were regulated by I942 without significant c-Jun recruitment, suggesting an additional, indirect mode of action for I942. In this regard, SOCS3 induction by I942 was found to require c-Jun and was associated with suppression of IL6-promoted ERK MAP kinase and AKT activity and induction of ICAM1. Pharmacological inhibition of ERK and AKT also potentiated ICAM1 induction by I942. We therefore propose that c-Jun activation by I942 regulates endothelial gene expression in HUVECs through direct mechanisms, involving recruitment of c-Jun or, as for ICAM1, through indirect regulation of tertiary regulators, including SOCS3.
Collapse
|
47
|
Shimizu A, Zankov DP, Kurokawa-Seo M, Ogita H. Vascular Endothelial Growth Factor-A Exerts Diverse Cellular Effects via Small G Proteins, Rho and Rap. Int J Mol Sci 2018; 19:ijms19041203. [PMID: 29659486 PMCID: PMC5979568 DOI: 10.3390/ijms19041203] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/03/2018] [Accepted: 04/12/2018] [Indexed: 12/18/2022] Open
Abstract
Vascular endothelial growth factors (VEGFs) include five molecules (VEGF-A, -B, -C, -D, and placental growth factor), and have various roles that crucially regulate cellular functions in many kinds of cells and tissues. Intracellular signal transduction induced by VEGFs has been extensively studied and is usually initiated by their binding to two classes of transmembrane receptors: receptor tyrosine kinase VEGF receptors (VEGF receptor-1, -2 and -3) and neuropilins (NRP1 and NRP2). In addition to many established results reported by other research groups, we have previously identified small G proteins, especially Ras homologue gene (Rho) and Ras-related protein (Rap), as important mediators of VEGF-A-stimulated signaling in cancer cells as well as endothelial cells. This review article describes the VEGF-A-induced signaling pathways underlying diverse cellular functions, including cell proliferation, migration, and angiogenesis, and the involvement of Rho, Rap, and their related molecules in these pathways.
Collapse
Affiliation(s)
- Akio Shimizu
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.
| | - Dimitar P Zankov
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.
| | - Misuzu Kurokawa-Seo
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan.
| | - Hisakazu Ogita
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.
| |
Collapse
|