1
|
Guo Q, Li J, Wang Z, Wu X, Jin Z, Zhu S, Li H, Zhang D, Hu W, Xu H, Yang L, Shi L, Wang Y. Potassium dehydroandrographolide succinate regulates the MyD88/CDH13 signaling pathway to enhance vascular injury-induced pathological vascular remodeling. Chin J Nat Med 2024; 22:62-74. [PMID: 38278560 DOI: 10.1016/s1875-5364(24)60562-5] [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: 07/23/2023] [Indexed: 01/28/2024]
Abstract
Pathological vascular remodeling is a hallmark of various vascular diseases. Previous research has established the significance of andrographolide in maintaining gastric vascular homeostasis and its pivotal role in modulating endothelial barrier dysfunction, which leads to pathological vascular remodeling. Potassium dehydroandrographolide succinate (PDA), a derivative of andrographolide, has been clinically utilized in the treatment of inflammatory diseases precipitated by viral infections. This study investigates the potential of PDA in regulating pathological vascular remodeling. The effect of PDA on vascular remodeling was assessed through the complete ligation of the carotid artery in C57BL/6 mice. Experimental approaches, including rat aortic primary smooth muscle cell culture, flow cytometry, bromodeoxyuridine (BrdU) incorporation assay, Boyden chamber cell migration assay, spheroid sprouting assay, and Matrigel-based tube formation assay, were employed to evaluate the influence of PDA on the proliferation and motility of smooth muscle cells (SMCs). Molecular docking simulations and co-immunoprecipitation assays were conducted to examine protein interactions. The results revealed that PDA exacerbates vascular injury-induced pathological remodeling, as evidenced by enhanced neointima formation. PDA treatment significantly increased the proliferation and migration of SMCs. Further mechanistic studies disclosed that PDA upregulated myeloid differentiation factor 88 (MyD88) expression in SMCs and interacted with T-cadherin (CDH13). This interaction augmented proliferation, migration, and extracellular matrix deposition, culminating in pathological vascular remodeling. Our findings underscore the critical role of PDA in the regulation of pathological vascular remodeling, mediated through the MyD88/CDH13 signaling pathway.
Collapse
Affiliation(s)
- Qiru Guo
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Jiali Li
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Zheng Wang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Xiao Wu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Zhong Jin
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Song Zhu
- Chengdu University of Traditional Chinese Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Hongfei Li
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Delai Zhang
- Chengdu University of Traditional Chinese Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Wangming Hu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Huan Xu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Lan Yang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Liangqin Shi
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
| | - Yong Wang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China.
| |
Collapse
|
2
|
Pei X, Li H, Yu H, Wang W, Mao D. APN Expression in Serum and Corpus Luteum: Regulation of Luteal Steroidogenesis Is Possibly Dependent on the AdipoR2/AMPK Pathway in Goats. Cells 2023; 12:1393. [PMID: 37408227 DOI: 10.3390/cells12101393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/23/2023] [Accepted: 05/09/2023] [Indexed: 07/07/2023] Open
Abstract
Adiponectin (APN) is an essential adipokine for a variety of reproductive processes. To investigate the role of APN in goat corpora lutea (CLs), CLs and sera from different luteal phases were collected for analysis. The results showed that the APN structure and content had no significant divergence in different luteal phases both in CLs and sera; however, high molecular weight APN was dominant in serum, while low molecular weight APN was more present in CLs. The luteal expression of both AdipoR1/2 and T-cadherin (T-Ca) increased on D11 and 17. APN and its receptors (AdipoR1/2 and T-Ca) were mainly expressed in goat luteal steroidogenic cells. The steroidogenesis and APN structure in pregnant CLs had a similar model as in the mid-cycle CLs. To further explore the effects and mechanisms of APN in CLs, steroidogenic cells from pregnant CLs were isolated to detect the AMPK-mediated pathway by the activation of APN (AdipoRon) and knockdown of APN receptors. The results revealed that P-AMPK in goat luteal cells increased after incubation with APN (1 μg/mL) or AdipoRon (25 μM) for 1 h, and progesterone (P4) and steroidogenic proteins levels (STAR/CYP11A1/HSD3B) decreased after 24 h. APN did not affect the steroidogenic protein expression when cells were pretreated with Compound C or SiAMPK. APN increased P-AMPK and reduced the CYP11A1 expression and P4 levels when cells were pretreated with SiAdipoR1 or SiT-Ca, while APN failed to affect P-AMPK, the CYP11A1 expression or the P4 levels when pretreated with SiAdipoR2. Therefore, the different structural forms of APN in CLs and sera may possess distinct functions; APN might regulate luteal steroidogenesis through AdipoR2 which is most likely dependent on AMPK.
Collapse
Affiliation(s)
- Xiaomeng Pei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Haolin Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Hao Yu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wei Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Dagan Mao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
3
|
Dasen B, Pigeot S, Born GM, Verrier S, Rivero O, Dittrich PS, Martin I, Filippova M. T-cadherin is a novel regulator of pericyte function during angiogenesis. Am J Physiol Cell Physiol 2023; 324:C821-C836. [PMID: 36802732 DOI: 10.1152/ajpcell.00326.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Pericytes are mural cells that play an important role in regulation of angiogenesis and endothelial function. Cadherins are a superfamily of adhesion molecules mediating Ca2+-dependent homophilic cell-cell interactions that control morphogenesis and tissue remodeling. To date, classical N-cadherin is the only cadherin described on pericytes. Here, we demonstrate that pericytes also express T-cadherin (H-cadherin, CDH13), an atypical glycosyl-phosphatidylinositol (GPI)-anchored member of the superfamily that has previously been implicated in regulation of neurite guidance, endothelial angiogenic behavior, and smooth muscle cell differentiation and progression of cardiovascular disease. The aim of the study was to investigate T-cadherin function in pericytes. Expression of T-cadherin in pericytes from different tissues was performed by immunofluorescence analysis. Using lentivirus-mediated gain-of-function and loss-of-function in cultured human pericytes, we demonstrate that T-cadherin regulates pericyte proliferation, migration, invasion, and interactions with endothelial cells during angiogenesis in vitro and in vivo. T-cadherin effects are associated with the reorganization of the cytoskeleton, modulation of cyclin D1, α-smooth muscle actin (αSMA), integrin β3, metalloprotease MMP1, and collagen expression levels, and involve Akt/GSK3β and ROCK intracellular signaling pathways. We also report the development of a novel multiwell 3-D microchannel slide for easy analysis of sprouting angiogenesis from a bioengineered microvessel in vitro. In conclusion, our data identify T-cadherin as a novel regulator of pericyte function and support that it is required for pericyte proliferation and invasion during active phase of angiogenesis, while T-cadherin loss shifts pericytes toward the myofibroblast state rendering them unable to control endothelial angiogenic behavior.
Collapse
Affiliation(s)
- Boris Dasen
- Tissue Engineering Lab, Department of Biomedicine and Department of Surgery, Basel University Hospital, Basel, Switzerland
| | - Sebastien Pigeot
- Tissue Engineering Lab, Department of Biomedicine and Department of Surgery, Basel University Hospital, Basel, Switzerland
| | - Gordian Manfred Born
- Tissue Engineering Lab, Department of Biomedicine and Department of Surgery, Basel University Hospital, Basel, Switzerland
| | | | - Olga Rivero
- Research Group on Psychiatry and Neurodegenerative Disorders, Biomedical Network Research Centre on Mental Health (CIBERSAM), Valencia, Spain
| | - Petra S Dittrich
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Ivan Martin
- Tissue Engineering Lab, Department of Biomedicine and Department of Surgery, Basel University Hospital, Basel, Switzerland
| | - Maria Filippova
- Tissue Engineering Lab, Department of Biomedicine and Department of Surgery, Basel University Hospital, Basel, Switzerland
| |
Collapse
|
4
|
Zhang D, Cui Y, Zhao M, Zheng X, Li C, Wei J, Wang K, Cui J. Orexin-A exerts neuroprotective effect in experimental intracerebral hemorrhage by suppressing autophagy via OXR1-mediated ERK/mTOR signaling pathway. Front Cell Neurosci 2022; 16:1045034. [PMID: 36619670 PMCID: PMC9815810 DOI: 10.3389/fncel.2022.1045034] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
Background Orexin-A (OXA) is a polypeptide produced in the hypothalamus, which binds to specific receptors and exerts multiple physiological effects. Autophagy plays a vital role in early brain injury (EBI) after intracerebral hemorrhage (ICH). However, the relationship between OXA and autophagy after ICH has not been confirmed. Methods In this study, the protective role of OXA was investigated in a model of hemin-induced injury in PC12 cells and blood-injection ICH model in rats, and its potential molecular mechanism was clarified. Neurobehavioral tests, brain water content, and pathologic morphology were assessed after ICH. Cell survival rate was determined using Cell Counting Kit-8 (CCK-8), while apoptosis was detected using flow cytometry. The autophagy protein LC3 that was originally identified as microtubule-associated protein 1 light 3 was evaluated by immunohistochemistry. The ultrastructural changes of cells following ICH were observed by transmission electron microscopy. Western blotting was performed to determine the expression levels of LC3, p62/SQSTM1 (p62), phosphorylated extracellular signal-regulated kinase 1/2 (p-ERK1/2), total extracellular signal-regulated kinase 1/2 (t-ERK1/2), mammalian target of rapamycin (mTOR), and phosphorylated mammalian target of rapamycin (p-mTOR). Results OXA treatment significantly improved neurofunctional outcomes, reduced brain edema, and alleviated neuronal apoptosis. OXA administration upregulated p-mTOR and p62, while it downregulated p-ERK1/2 and LC3; this effect was reversed by the orexin receptor 1 (OXR1) antagonist SB-334867. Conclusions This study demonstrates that OXA suppresses autophagy via the OXR1-mediated ERK/mTOR signaling pathway to exert neuroprotective effects, and it might provide a novel therapeutic approach in patients suffering from ICH.
Collapse
Affiliation(s)
- Dexin Zhang
- Department of Surgery, Hebei Medical University, Shijiazhuang, China
| | - Ying Cui
- Department of Neurology, Tangshan Gongren Hospital, Tangshan, China
| | - Manman Zhao
- Department of Histology and Embryology, North China University of Science and Technology, Tangshan, China
| | - Xuecheng Zheng
- Department of Surgery, Hebei Medical University, Shijiazhuang, China
| | - Chunyan Li
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jingbo Wei
- Department of Histology and Embryology, North China University of Science and Technology, Tangshan, China
| | - Kaijie Wang
- Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, China
| | - Jianzhong Cui
- Department of Surgery, Hebei Medical University, Shijiazhuang, China,Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, China,*Correspondence: Jianzhong Cui,
| |
Collapse
|
5
|
Wang S, Yuan R, Liu M, Zhang Y, Jia B, Ruan J, Shen J, Zhang Y, Liu M, Wang T. Targeting autophagy in atherosclerosis: Advances and therapeutic potential of natural bioactive compounds from herbal medicines and natural products. Biomed Pharmacother 2022; 155:113712. [PMID: 36130420 DOI: 10.1016/j.biopha.2022.113712] [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: 06/29/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 11/29/2022] Open
Abstract
Atherosclerosis (AS) is the most common causes of cardiovascular disease characterized by the formation of atherosclerotic plaques in the arterial wall, and it has become a dominant public health problem that seriously threaten people worldwide. Autophagy is a cellular self-catabolism process, which is critical to protect cellular homeostasis against harmful conditions. Emerging evidence suggest that dysregulated autophagy is involved in the development of AS. Therefore, pharmacological interventions have been developed to inhibit the AS via autophagy induction. Among various AS treating methods, herbal medicines and natural products have been applied as effective complementary and alternative medicines to ameliorate AS and its associated cardiovascular disease. Recently, mounting evidence revealed that natural bioactive compounds from herbs and natural products could induce autophagy to suppress the occurrence and development of AS, by promoting cholesterol efflux, reducing plaque inflammation, and inhibiting apoptosis or senescence. In the present review, we highlight recent findings regarding possible effects and molecular mechanism of natural compounds in autophagy-targeted mitigation of atherosclerosis, aiming to provide new potential therapeutic strategies for the atherosclerosis treatment preclinically and clinically.
Collapse
Affiliation(s)
- Sijian Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ruolan Yuan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Miao Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yiwen Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bona Jia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jingya Ruan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiayan Shen
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Mengyang Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Tao Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
6
|
Zou Y, Chen Z, Zhang X, Yu J, Xu H, Cui J, Li Y, Niu Y, Zhou C, Xia J, Wu J. Targeting PCSK9 Ameliorates Graft Vascular Disease in Mice by Inhibiting NLRP3 Inflammasome Activation in Vascular Smooth Muscle Cells. Front Immunol 2022; 13:894789. [PMID: 35720337 PMCID: PMC9204514 DOI: 10.3389/fimmu.2022.894789] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/28/2022] [Indexed: 12/23/2022] Open
Abstract
Background Graft vascular disease (GVD), which limits the long-term survival of patients after solid-organ transplantation, is associated with both immune responses and nonimmune factors, including dyslipidemia. Recent studies have shown that inhibition of proprotein convertase subtilisin/kexin type 9 (PCSK9), a U.S. Federal Drug Administration-approved treatment for hyperlipidemia, reduces cardiovascular events, regulates inflammatory responses, and enhances the efficacy of immune checkpoint therapy in cancer treatment through a cholesterol-independent mechanism. However, whether targeting PCSK9 is a potential therapeutic strategy for GVD remains unknown. Methods Serum samples and grafts were harvested from male mice undergoing abdominal aortic transplantation. The pathological alterations in the aortic grafts were detected by hematoxylin and eosin staining, Verhoeff’s Van Gieson staining, and Masson staining. Inflammatory cell infiltration and proinflammatory cytokine expression in the aortic grafts were detected by immunohistochemistry and quantitative real-time polymerase chain reaction (qRT-PCR), respectively. The regulatory effects of PCSK9 on vascular smooth muscle cell (VSMC) migration and proliferation were examined by transwell, EdU, and western blot assays. The effect of Evolocumab, a PCSK9 inhibitor, on GVD in humanized PCSK9 mice was also evaluated. Results PCSK9 was upregulated in the serum, grafts, and liver of mice in the allograft group subjected to abdominal aortic transplantation. Pcsk9 knockout significantly reduced vascular stenosis, the intimal hyperplasia area and collagen deposition. Pcsk9 depletion also inhibited macrophage recruitment and the mRNA expression of proinflammatory cytokines in aortic grafts. Furthermore, Pcsk9 knockout suppressed the migration and proliferation of VSMCs, which was related to the inhibition of NLRP3 inflammasome activation. Meanwhile, Evolocumab significantly ameliorated GVD in humanized PCSK9 mice. Conclusion PCSK9 is upregulated in a mouse model of GVD, and Pcsk9 knockout reduces vascular occlusion, suggesting that PCSK9 may be a promising target for the treatment of GVD.
Collapse
Affiliation(s)
- Yanqiang Zou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhang Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jikai Cui
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqing Niu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Ravasio A, Morselli E, Bertocchi C. Mechanoautophagy: Synergies Between Autophagy and Cell Mechanotransduction at Adhesive Complexes. Front Cell Dev Biol 2022; 10:917662. [PMID: 35721483 PMCID: PMC9198486 DOI: 10.3389/fcell.2022.917662] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/13/2022] [Indexed: 12/15/2022] Open
Abstract
Cells are exposed and respond to various mechanical forces and physical cues stemming from their environment. This interaction has been seen to differentially regulate various cellular processes for maintenance of homeostasis, of which autophagy represents one of the major players. In addition, autophagy has been suggested to regulate mechanical functions of the cells including their interaction with the environment. In this minireview, we summarize the state of the art of the fascinating interplay between autophagy and the mechanotransduction machinery associated with cell adhesions, that we name ¨Mechanoautophagy¨
Collapse
Affiliation(s)
- Andrea Ravasio
- Institute for Biological and Medical Engineering Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- *Correspondence: Cristina Bertocchi, ; Andrea Ravasio,
| | - Eugenia Morselli
- Department of Basic Sciences, Faculty of Medicine and Sciences, Universidad San Sebastián, Santiago, Chile
| | - Cristina Bertocchi
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology Pontificia Universidad Católica de Chile, Santiago, Chile
- *Correspondence: Cristina Bertocchi, ; Andrea Ravasio,
| |
Collapse
|
8
|
Chignon A, Mathieu S, Rufiange A, Argaud D, Voisine P, Bossé Y, Arsenault BJ, Thériault S, Mathieu P. Enhancer promoter interactome and Mendelian randomization identify network of druggable vascular genes in coronary artery disease. Hum Genomics 2022; 16:8. [PMID: 35246263 PMCID: PMC8895522 DOI: 10.1186/s40246-022-00381-4] [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: 10/22/2021] [Accepted: 02/17/2022] [Indexed: 11/14/2022] Open
Abstract
Coronary artery disease (CAD) is a multifactorial disorder, which is partly heritable. Herein, we implemented a mapping of CAD-associated candidate genes by using genome-wide enhancer-promoter conformation (H3K27ac-HiChIP) and expression quantitative trait loci (eQTL). Enhancer-promoter anchor loops from human coronary artery smooth muscle cells (HCASMC) explained 22% of the heritability for CAD. 3D enhancer-promoter genome mapping of CAD-genes in HCASMC was enriched in vascular eQTL genes. By using colocalization and Mendelian randomization analyses, we identified 58 causal candidate vascular genes including some druggable targets (MAP3K11, CAMK1D, PDGFD, IPO9 and CETP). A network analysis of causal candidate genes was enriched in TGF beta and MAPK pathways. The pharmacologic inhibition of causal candidate gene MAP3K11 in vascular SMC reduced the expression of athero-relevant genes and lowered cell migration, a cardinal process in CAD. Genes connected to enhancers are enriched in vascular eQTL and druggable genes causally associated with CAD.
Collapse
Affiliation(s)
- Arnaud Chignon
- Laboratory of Cardiovascular Pathobiology, Department of Surgery, Institut de Cardiologie Et de Pneumologie de Québec, Quebec Heart and Lung Institute/Research Center, Laval University, 2725 Chemin Ste-Foy, Québec, QC, G1V-4G5, Canada
| | - Samuel Mathieu
- Laboratory of Cardiovascular Pathobiology, Department of Surgery, Institut de Cardiologie Et de Pneumologie de Québec, Quebec Heart and Lung Institute/Research Center, Laval University, 2725 Chemin Ste-Foy, Québec, QC, G1V-4G5, Canada
| | - Anne Rufiange
- Laboratory of Cardiovascular Pathobiology, Department of Surgery, Institut de Cardiologie Et de Pneumologie de Québec, Quebec Heart and Lung Institute/Research Center, Laval University, 2725 Chemin Ste-Foy, Québec, QC, G1V-4G5, Canada
| | - Déborah Argaud
- Laboratory of Cardiovascular Pathobiology, Department of Surgery, Institut de Cardiologie Et de Pneumologie de Québec, Quebec Heart and Lung Institute/Research Center, Laval University, 2725 Chemin Ste-Foy, Québec, QC, G1V-4G5, Canada
| | | | - Yohan Bossé
- Department of Molecular Medicine, Laval University, Quebec, Canada
| | | | - Sébastien Thériault
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Quebec, Canada
| | - Patrick Mathieu
- Laboratory of Cardiovascular Pathobiology, Department of Surgery, Institut de Cardiologie Et de Pneumologie de Québec, Quebec Heart and Lung Institute/Research Center, Laval University, 2725 Chemin Ste-Foy, Québec, QC, G1V-4G5, Canada. .,Department of Surgery, Laval University, Quebec, Canada.
| |
Collapse
|
9
|
Kobelt D, Perez-Hernandez D, Fleuter C, Dahlmann M, Zincke F, Smith J, Migotti R, Popp O, Burock S, Walther W, Dittmar G, Mertins P, Stein U. The newly identified MEK1 tyrosine phosphorylation target MACC1 is druggable by approved MEK1 inhibitors to restrict colorectal cancer metastasis. Oncogene 2021; 40:5286-5301. [PMID: 34247190 PMCID: PMC8390371 DOI: 10.1038/s41388-021-01917-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/14/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023]
Abstract
Cancer metastasis causes >90% of cancer deaths and remains a major treatment challenge. Here we deciphered the impact of tyrosine phosphorylation of MACC1, a causative driver for cancer metastasis, for cancer cell signaling and novel interventions to restrict cancer metastasis. We identified MACC1 as new MEK1 substrate. MEK1 directly phosphorylates MACC1, leading to accelerated and increased ERK1 activation. Mutating in silico predicted hierarchical MACC1 tyrosine phosphorylation sites abrogates MACC1-induced migration, invasion, and MET expression, a transcriptional MACC1 target. Targeting MEK1 by RNAi or clinically applicable MEK1 inhibitors AZD6244 and GSK1120212 reduces MACC1 tyrosine phosphorylation and restricts MACC1-induced metastasis formation in mice. Although MEK1 levels, contrary to MACC1, are not of prognostic relevance for CRC patients, MEK1 expression was found indispensable for MACC1-induced metastasis. This study identifies MACC1 as new MEK1 substrate for tyrosine phosphorylation decisively impacting cell motility, tumor growth, and metastasis. Thus, MAP kinase signaling is not linear leading to ERK activation, but branches at the level of MEK1. This fundamental finding opens new therapeutic options for targeting the MEK1/MACC1 axis as novel vulnerability in patients at high risk for metastasis. This might be extended from CRC to further solid tumor entities.
Collapse
Affiliation(s)
- Dennis Kobelt
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Daniel Perez-Hernandez
- Mass Spectrometry Core Unit, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Proteome and Genome Research Laboratory, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Claudia Fleuter
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Mathias Dahlmann
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Fabian Zincke
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Janice Smith
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Rebekka Migotti
- Mass Spectrometry Core Unit, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Oliver Popp
- Mass Spectrometry Core Unit, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Susen Burock
- Charité Comprehensive Cancer Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Wolfgang Walther
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Gunnar Dittmar
- Mass Spectrometry Core Unit, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Proteome and Genome Research Laboratory, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Philipp Mertins
- Mass Spectrometry Core Unit, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Ulrike Stein
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
10
|
Autophagy-Related Genes in Atherosclerosis. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:6402206. [PMID: 34306596 PMCID: PMC8270709 DOI: 10.1155/2021/6402206] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/02/2021] [Accepted: 06/22/2021] [Indexed: 01/22/2023]
Abstract
Background Atherosclerosis (AS) is a common chronic vascular inflammatory disease and one of the main causes of cardiovascular/cerebrovascular diseases (CVDs). Autophagy-related genes (ARGs) play a crucial part in pathophysiological processes of AS. However, the expression profile of ARGs has rarely been adopted to explore the relationship between autophagy and AS. Therefore, using the expression profile of ARGs to explore the relationship between autophagy and AS may provide new insights for the treatment of CVDs. Methods The differentially expressed ARGs of the GSE57691 dataset were obtained from the Human Autophagy Database (HADb) and the Gene Expression Omnibus (GEO) database, and the GSE57691 dataset contains 9 aortic atheroma tissues and 10 normal aortic tissues. The differentially expressed ARGs of the GSE57691 dataset were analyzed by protein-protein interaction (PPI), gene ontology analysis (GO), and Kyoto Encyclopedia of Genes and Genomes analysis (KEGG) and were chosen to explore related miRNAs/transcriptional factors. Results The GSE57691 dataset had a total of 41 differentially expressed ARGs. The GO analysis results revealed that ARGs were mainly enriched in autophagy, autophagosome, and protein serine/threonine kinase activity. KEGG analysis results showed that ARGs were mainly enriched in autophagy-animal and longevity regulating signaling pathways. Expressions of ATG5, MAP1LC3B, MAPK3, MAPK8, and RB1CC1 were regarded as focus in the PPI regulatory networks. Furthermore, 11 related miRNAs and 6 related transcription factors were obtained by miRNAs/transcription factor target network analysis. Conclusions Autophagy and ARGs may play a vital role in regulating the pathophysiology of AS.
Collapse
|
11
|
Lu Q, Huang Y, Wu J, Guan Y, Du M, Wang F, Liu Z, Zhu Y, Gong G, Hou H, Zhang M, Zhang JY, Ning F, Chen L, Wang L, Lash GE. T-cadherin inhibits invasion and migration of endometrial stromal cells in endometriosis. Hum Reprod 2021; 35:145-156. [PMID: 31886853 DOI: 10.1093/humrep/dez252] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 09/10/2019] [Indexed: 12/18/2022] Open
Abstract
STUDY QUESTION What is the expression level of T-cadherin in endometriosis, and does T-cadherin play a role in regulating invasion and migration of endometrial stromal cells? SUMMARY ANSWER T-cadherin expression was reduced in ectopic endometriotic lesions compared to eutopic endometrium, and T-cadherin overexpression inhibited the invasion and migration of endometrial stromal cells. WHAT IS KNOWN ALREADY Endometriosis is a disease that involves active cell invasion and migration. T-cadherin can inhibit cell invasion, migration and proliferation in various cancer cells, but its role in endometriosis has not been investigated. STUDY DESIGN, SIZE, DURATION We explored the expression status of T-cadherin in 40 patients with and 24 without endometriosis. We also isolated endometrial stromal cells to study the invasion, migration and signaling pathway regulation of T-cadherin overexpression. PARTICIPANTS/MATERIALS, SETTING, METHODS Patients were recruited at the Guangzhou Women and Children's Medical Center to study the expression levels of T-cadherin. The expression of T-cadherin was detected by immunohistochemistry staining and western blot. H-score was used to evaluate the staining intensity of T-cadherin. The correlation between T-cadherin expression levels (H-score) and endometriosis patients' age, stage, lesion size and adhesion was analyzed. Endometrial stromal cells from patients with and without endometriosis were isolated, and cell invasion and migration were detected by transwell assays after T-cadherin overexpression. The expression of vimentin in T-cadherin-overexpressed cells was detected by western blot. After T-cadherin overexpression, the phosphorylation profile of signaling pathway proteins was detected with the Proteome Profiler Human Phospho-Kinase Array Kit. MAIN RESULTS AND THE ROLE OF CHANCE There was no difference in the expression of T-cadherin in the normal endometrium of control patients and the eutopic endometrium of endometriotic patients, but it was significantly decreased in the ectopic endometrium of endometriotic patients, compared with control endometrium and eutopic endometrium of endometriosis patients (P < 0.0001, for both). Western blot analysis also showed that the expression of T-cadherin was decreased in ectopic endometriotic lesions, but not the normal control endometrium or the endometriotic eutopic endometrium. The results of transwell assays indicated that T-cadherin overexpression inhibited the invasion and migration of endometrial stromal cells. In addition, T-cadherin overexpression promoted the phosphorylation of HSP27 (S78/S82) and JNK 1/2/3 (T183/Y185, T221/Y223) and decreased the expression of vimentin, MMP2 and MMP9 in eutopic endometriosis stromal cells. LARGE-SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION The control group were patients with benign gynecological conditions (e.g. uterus myoma, endometrial or cervical polyp), which may have genetic or epigenetic variations associated with T-cadherin expression and signaling pathways. The case numbers of involved endometriosis and control patients were limited. This study only used endometrial stromal cells from patients with or without endometriosis. Ideally, ectopic endometrial stromal cells of the ovarian endometriotic lesions should also be utilized to explore the function of T-cadherin. WIDER IMPLICATIONS OF THE FINDINGS Further investigation of the role of T-cadherin in endometriosis may generate new potential therapeutic targets for this complex disorder. STUDY FUNDING AND COMPETING INTEREST(S) This study was supported by the Natural Science Foundation of Guangdong Province (2016A030313495), National Natural Science Foundation of China (81702567, 81671406, 31871412), the Science and Technology Programs of Guangdong (2017A050501021), Medical Science Technology Research Fund of Guangdong Province (A2018075), the Science and Technology Programs of Guangzhou City (201704030103), Internal Project of Family Planning Research Institute of Guangdong Province (S2018004), Post-doc initiation fund of Guangzhou (3302) and Post-doc science research initiation fund of Guangzhou Women and Children's Medical Center (20160322). There are no conflicts of interest.
Collapse
Affiliation(s)
- Qinsheng Lu
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, P.R. China
| | - Yanqing Huang
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, P.R. China
| | - Jiabao Wu
- NHC Key Laboratory of Male Reproduction and Genetics, Family Planning Research Institute of Guangdong Province, Guangzhou, Guangdong 510600, P.R. China
| | - Yutao Guan
- Department of Obstetrics and Gynecology, the First People's Hospital of Foshan, Foshan, Guangdong 528000, P.R. China
| | - Miaomiao Du
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, P.R. China
| | - Fenghua Wang
- Department of Pathology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| | - Zhihong Liu
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, P.R. China
| | - Yali Zhu
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, P.R. China
| | - Guifang Gong
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, P.R. China
| | - Huomei Hou
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, P.R. China
| | - Min Zhang
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, P.R. China
| | - Joy Yue Zhang
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, P.R. China
| | - Fen Ning
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, P.R. China
| | - Lixin Chen
- Department of Physiology, Medical College, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Liwei Wang
- Department of Physiology, Medical College, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Gendie E Lash
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, P.R. China
| |
Collapse
|
12
|
Orexin-A protects against cerebral ischemia-reperfusion injury by inhibiting excessive autophagy through OX1R-mediated MAPK/ERK/mTOR pathway. Cell Signal 2020; 79:109839. [PMID: 33212156 DOI: 10.1016/j.cellsig.2020.109839] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/13/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023]
Abstract
Orexin A (OXA) is a neuroprotective peptide that exerts protective effects on multiple physiological and pathological processes. Activation of autophagy is linked to the occurrence of cerebral ischemia-reperfusion injury (CIRI); however, its function remains incompletely understood. In this study, OXA was sought to exert its neuroprotective role by regulating autophagy in oxygen and glucose deprivation and reoxygenation (OGD/R) model and middle cerebral artery occlusion (MCAO) model of rats, and to elucidate the underlying molecular mechanisms. Acridine orange (AO) staining was used to evaluate autophagic vacuoles. Cell viability was measured by CCK8. The levels of p-ERK1/2, t-ERK1/2, p-mTOR, LC3B, Beclin 1, and p62 were evaluated by western blotting. Apoptosis rate was detected by Hoechst 33342 staining and Terminal deoxynucleotidyltransferase-mediated dUTP nick-end labeling (TUNEL). OXA treatment alleviated neuronal apoptosis and significantly inhibited autophagy activity. Mechanistically, OXA exerted its neuroprotective effects in vivo and in vitro by suppressing over-activated autophagy by modulating OX1R-mediated MAPK/ERK/mTOR pathway. The results of this study elucidate the roles of autophagy in CIRI and the mechanisms underlying the neuroprotective action of OXA. Our findings could facilitate the development of novel therapeutics for ischemic stroke.
Collapse
|
13
|
Zhang YY, Shi YN, Zhu N, Wang W, Deng CF, Xie XJ, Liao DF, Qin L. Autophagy: a killer or guardian of vascular smooth muscle cells. J Drug Target 2020; 28:449-455. [DOI: 10.1080/1061186x.2019.1705312] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Yin-Yu Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
- Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, China
| | - Ya-Ning Shi
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
- Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, China
| | - Neng Zhu
- The First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, China
| | - Wei Wang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
- Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, China
| | - Chang-Feng Deng
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
- Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, China
| | - Xue-Jiao Xie
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Duan-Fang Liao
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
- Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, China
| | - Li Qin
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
- Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
14
|
Li HY, Hong X, Cao QQ, So KF. Adiponectin, exercise and eye diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 147:281-294. [PMID: 31607358 DOI: 10.1016/bs.irn.2019.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adiponectin, one kind of adipokines, has been shown to be neuroprotective in different neurodegenerative diseases. Adiponectin exerts its role through combination with its receptors and activates downstream molecular pathways. In the retinas, the expression of adiponectin can be detected and adiponectin receptors (AdipoRs) locate in different retinal cells. Adiponectin is mainly produced by adipose tissue, enters the circulation and passes through blood-brain barrier (BBB) without injury. It can also be produced locally in the brains as well as in the retinas. Therefore, it is possible that adiponectin from blood as well as that produced locally in the retinas take part in defense of different eye diseases. Here we have summarized the published data about the protective effects of adiponectin in eye diseases. Because exercise can increase the production of adiponectin systemically in the whole body and locally in the brain although no evidence has shown that exercise can increase the production of adiponectin in the eyes until now, we hypothesize that exercise will have a potential protective effect for the eyes via increasing the levels of adiponectin which needs further investigation.
Collapse
Affiliation(s)
- Hong-Ying Li
- Department of Anatomy, Medical School, Jinan University, Guangzhou, PR China; Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, PR China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, PR China.
| | - Xi Hong
- Department of Anatomy, Medical School, Jinan University, Guangzhou, PR China
| | - Qian-Qian Cao
- Department of Anatomy, Medical School, Jinan University, Guangzhou, PR China
| | - Kwok-Fai So
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, PR China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, PR China; Guangdong Key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou, PR China; State Key Laboratory of Brain and Cognitive Sciences and Department of Ophthalmology, The University of HongKong, Hong Kong, PR China.
| |
Collapse
|
15
|
Hou B, Wang G, Gao Q, Wei Y, Zhang C, Wang Y, Huo Y, Yang H, Jiang X, Xi Z. SQSTM1/p62 loss reverses the inhibitory effect of sunitinib on autophagy independent of AMPK signaling. Sci Rep 2019; 9:11087. [PMID: 31366950 PMCID: PMC6668422 DOI: 10.1038/s41598-019-47597-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 07/19/2019] [Indexed: 02/03/2023] Open
Abstract
Sunitinib (ST), a multitargeted receptor tyrosine kinase inhibitor, has been demonstrated to be effective for the treatment of renal carcinoma. It has been reported that ST is involved in the mediation of autophagy; however, its regulatory role in the autophagic process remains controversial. Furthermore, the mechanism by which activated AMP-activated protein kinase (AMPK) negatively regulates autophagy remains nearly unexplored. In the present study, we revealed that ST inhibited AMPK activity and regulated autophagy in a cell type- and dose-dependent manner. In a number of cell lines, ST was demonstrated to inhibit H2O2-induced autophagy and the phosphorylation of acetyl-CoA carboxylase (ACC), whereas alone it could block the autophagic flux concurrent with increased expression of p62. An immunoprecipitation assay revealed that LC3 directly interacted with p62, whereas ST increased punctate LC3 staining, which was well colocalized with p62. Taken together, we reveal a previously unnoticed pathway for ST to regulate the autophagic process, and p62, although often utilized as a substrate in autophagy, plays a critical role in regulating the inhibition of ST in both basal and induced autophagy.
Collapse
Affiliation(s)
- Bolin Hou
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Gang Wang
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
| | - Quan Gao
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Yanjie Wei
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Caining Zhang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Yange Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Yuqing Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, 30912, Georgia, USA
| | - Huaiyi Yang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xuejun Jiang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Zhijun Xi
- Department of Urology, Peking University First Hospital, Beijing, 100034, China.
| |
Collapse
|
16
|
Ba L, Gao J, Chen Y, Qi H, Dong C, Pan H, Zhang Q, Shi P, Song C, Guan X, Cao Y, Sun H. Allicin attenuates pathological cardiac hypertrophy by inhibiting autophagy via activation of PI3K/Akt/mTOR and MAPK/ERK/mTOR signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 58:152765. [PMID: 31005720 DOI: 10.1016/j.phymed.2018.11.025] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 11/16/2018] [Accepted: 11/17/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Cardiac hypertrophy is an adaptive response of the myocardium to pressure or volume overload. Recent evidences indicate that allicin can prevent cardiac hypertrophy. However, it is not clear whether allicin alleviates cardiac hypertrophy by inhibiting autophagy. PURPOSE We aimed to investigate the effects of allicin on pressure overload-induced cardiac hypertrophy, and further to clarify the related mechanism. STUDY DESIGN/METHODS Cardiac hypertrophy was successfully established by abdominal aortic constriction (AAC) in rats, and cardiomyocytes hypertrophy was simulated by angiotensin II (Ang II) in vitro. Hemodynamic parameters were monitored by organism function experiment system in vivo. The changes of cell surface area were observed using HE and immunofluorescence staining in vivoand in vitro, respectively. The expressions of cardiac hypertrophy relative protein (BNP and β-MHC), autophagy marker protein (LC3-II and Beclin-1), Akt, PI3K and ERK were detected by western blot. RESULTS Allicin could improve cardiac function, and reduce cardiomyocytes size, and decrease BNP and β-MHC protein expressions. Further results showed that allicin could lower LC3-II and Beclin-1 protein expressions both in vivo and in vitro experiments. And pharmacological inhibitor of mTOR, rapamycin could antagonize the effects of allicin on Ang II-induced cardiac hypertrophy and autophagy. Simultaneously, allicin could promote the expressions of p-Akt, p-PI3K and p-ERK protein. CONCLUSION These findings reveal a novel mechanism of allicin attenuating cardiac hypertrophy which allicin could inhibit excessive autophagy via activating PI3K/Akt/mTOR and MAPK/ERK/mTOR signaling pathways.
Collapse
Affiliation(s)
- Lina Ba
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Jingquan Gao
- Department of Clinical Nursing, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Yunping Chen
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Hanping Qi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Chonghui Dong
- Department of Pharmacy, Fifth Clinical College of Harbin Medical University, Daqing, Heilongjiang 163316, China
| | - Hao Pan
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Qianhui Zhang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Pilong Shi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Chao Song
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Xueying Guan
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Yonggang Cao
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China.
| | - Hongli Sun
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China.
| |
Collapse
|
17
|
Chen Q, Gao R, Geng Y, Chen X, Liu X, Zhang L, Mu X, Ding Y, Wang Y, He J. Decreased autophagy was implicated in the decreased apoptosis during decidualization in early pregnant mice. J Mol Histol 2018; 49:589-597. [PMID: 30298448 DOI: 10.1007/s10735-018-9797-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/26/2018] [Indexed: 11/25/2022]
Abstract
Folate deficiency is a major risk factor of birth defects. Mechanistic studies on folate deficiency resulting in birth defects have mainly focused on fetal development. There have been few studies on folate deficiency from the point of view of the mother's uterus. In our previous study, we demonstrated that folate deficiency inhibits apoptosis of decidual cells, thereby restraining decidualization of the endometrium and impairing pregnancy. In this study, we further investigated the potential mechanism by which folate deficiency decreases endometrial apoptosis during decidualization. To investigate whether endometrium autophagy was inhibited under folate deficiency during decidualization, we performed real-time PCR for endometrial LC3 and P62 on day 6 (D6) to D8 of pregnancy in mice, and both were significantly changed compared to non-folate-deficient mice. Western blots showed that LC3-II and P62 were also changed in folate-deficient mice. Compared with control mice, a few punctuate LC3-II structures were detected in the folate deficiency group by immunofluorescence. Transmission electron micrographs of decidual cells on D8 showed that there were no evident autophagosomes in the folate deficiency group. In addition, apoptosis-related protein analysis by western blotting, TUNEL staining and flow cytometry showed that decreased endometrial apoptosis on D8 of pregnancy under folate deficiency was reversed after treatment with rapamycin, an autophagy inducer. ROS measurement showed that the endometrium ROS level was reduced by folate deficiency and that rapamycin reversed this effect on day 8 of pregnancy. All the results suggest that inhibiting endometrial autophagy may be implicated in the decreased endometrial apoptosis under folate deficiency during decidualization.
Collapse
Affiliation(s)
- Qiutong Chen
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Box 197, Chongqing, 400016, People's Republic of China
| | - Rufei Gao
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Box 197, Chongqing, 400016, People's Republic of China
| | - Yanqing Geng
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Box 197, Chongqing, 400016, People's Republic of China
| | - Xuemei Chen
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Box 197, Chongqing, 400016, People's Republic of China
| | - Xueqing Liu
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Box 197, Chongqing, 400016, People's Republic of China
| | - Lei Zhang
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Box 197, Chongqing, 400016, People's Republic of China
| | - Xinyi Mu
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Box 197, Chongqing, 400016, People's Republic of China
| | - Yubin Ding
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Box 197, Chongqing, 400016, People's Republic of China
| | - Yingxiong Wang
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Box 197, Chongqing, 400016, People's Republic of China
| | - Junlin He
- Laboratory of Reproductive Biology, School of Public Health and Management, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Box 197, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
18
|
Gao P, Wu W, Ye J, Lu YW, Adam AP, Singer HA, Long X. Transforming growth factor β1 suppresses proinflammatory gene program independent of its regulation on vascular smooth muscle differentiation and autophagy. Cell Signal 2018; 50:160-170. [PMID: 30006123 DOI: 10.1016/j.cellsig.2018.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/19/2018] [Accepted: 07/09/2018] [Indexed: 01/01/2023]
Abstract
Transforming growth factor β (TGFβ) signaling plays crucial roles in maintaining vascular integrity and homeostasis, and is established as a strong activator of vascular smooth muscle cell (VSMC) differentiation. Chronic inflammation is a hallmark of various vascular diseases. Although TGFβ signaling has been suggested to be protective against inflammatory aortic aneurysm progression, its exact effects on VSMC inflammatory process and the underlying mechanisms are not fully unraveled. Here we revealed that TGFβ1 suppressed the expression of a broad array of proinflammatory genes while potently induced the expression of contractile genes in cultured primary human coronary artery SMCs (HCASMCs). The regulation of TGFβ1 on VSMC contractile and proinflammatory gene programs appeared to occur in parallel and both processes were through a SMAD4-dependent canonical pathway. We also showed evidence that the suppression of TGFβ1 on VSMC proinflammatory genes was mediated, at least partially through the blockade of signal transducer and activator of transcription 3 (STAT3) and NF-κB pathways. Interestingly, our RNA-seq data also revealed that TGFβ1 suppressed gene expression of a battery of autophagy mediators, which was validated by western blot for the conversion of microtubule-associated protein light chain 3 (LC3) and by immunofluo-rescence staining for LC3 puncta. However, impairment of VSMC autophagy by ATG5 deletion failed to rescue TGFβ1 influence on both VSMC contractile and proinflammatory gene programs, suggesting that TGFβ1-regulated VSMC differentiation and inflammation are not attributed to TGFβ1 suppression on autophagy. In summary, our results demonstrated an important role of TGFβ signaling in suppressing proinflammatory gene program in cultured primary human VSMCs via the blockade on STAT3 and NF-κB pathway, therefore providing novel insights into the mechanisms underlying the protective role of TGFβ signaling in vascular diseases.
Collapse
Affiliation(s)
- Ping Gao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Wen Wu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Jiemei Ye
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Yao Wei Lu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Alejandro Pablo Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States; Department of Ophthalmology, Albany Medical College, Albany, NY, United States
| | - Harold A Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Xiaochun Long
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States.
| |
Collapse
|
19
|
Frismantiene A, Philippova M, Erne P, Resink TJ. Cadherins in vascular smooth muscle cell (patho)biology: Quid nos scimus? Cell Signal 2018; 45:23-42. [DOI: 10.1016/j.cellsig.2018.01.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/23/2018] [Accepted: 01/23/2018] [Indexed: 12/16/2022]
|
20
|
Cao X, Han C, Wen J, Guo X, Zhang K. Nicotine increases apoptosis in HUVECs cultured in high glucose/high fat via Akt ubiquitination and degradation. Clin Exp Pharmacol Physiol 2017; 45:198-204. [PMID: 28963785 DOI: 10.1111/1440-1681.12865] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/04/2017] [Accepted: 09/19/2017] [Indexed: 12/12/2022]
Abstract
It is well-documented that nicotine, the main active ingredient in cigarettes, results in endothelial cell injury in numerous diseases. However, whether nicotine plays a crucial role in endothelial cell injury in diabetes and the exact molecular mechanism that mediates this process have not been fully elucidated. The current study aimed to investigate the effects of nicotine on endothelial cell injury in diabetes and the specific molecular mechanism by which it plays a role. Human umbilical vein endothelial cells (HUVECs) were incubated in HG/HF media and treated with nicotine, PYR-41 (a selective ubiquitin E1 inhibitor), Akt-overexpressing adenovirus, or TTC3 and MUL1 shRNA adenovirus. Cell viability was subsequently detected by the CCK8 assay, and apoptosis was examined by caspase-3 cleavage and activity analysis. Compared to the HG/HF incubated group, nicotine incubation significantly decreased cell survival and increased apoptosis. Moreover, nicotine induced Akt degradation via UPS, and Akt overexpression blocked nicotine-induced apoptosis in HUVECs cultured in HG/HF media. Furthermore, the TTC3 and MUL1 shRNA adenovirus dramatically decreased the Akt ubiquitination and apoptosis induced by nicotine. These results indicate that nicotine-induced Akt ubiquitination and degradation occurs through TTC3 and MUL1 and results in a dramatic increase in apoptosis in HUVECs cultured in HG/HF media.
Collapse
Affiliation(s)
- Xiaofang Cao
- Center for Disease Control and Prevention of Baoji City, Baoji City, Shanxi Province, China
| | - Chunling Han
- Baoji Maternal and Child Health Hospital, Baoji City, Shanxi Province, China
| | - Jinsuo Wen
- Center for Disease Control and Prevention of Baoji City, Baoji City, Shanxi Province, China
| | - Xiaokun Guo
- Center for Disease Control and Prevention of Baoji City, Baoji City, Shanxi Province, China
| | - Kejian Zhang
- Center for Disease Control and Prevention of Baoji City, Baoji City, Shanxi Province, China
| |
Collapse
|
21
|
Halper J. Basic Components of Vascular Connective Tissue and Extracellular Matrix. ADVANCES IN PHARMACOLOGY 2017; 81:95-127. [PMID: 29310805 DOI: 10.1016/bs.apha.2017.08.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Though the composition of the three layers constituting the blood vessel wall varies among the different types of blood vessels, and some layers may even be missing in capillaries, certain basic components, and properties are shared by all blood vessels, though each histologically distinct layer contains a unique complement of extracellular components, growth factors and cytokines, and cell types as well. The structure and composition of vessel layers informs and is informed by the function of the particular blood vessel. The adaptation of the composition and the resulting function of the extracellular matrix (ECM) to changes in circulation/blood flow and a variety of other extravascular stimuli can be characterized as remodeling spearheaded by vascular cells. There is a surprising amount of cell traffic among the three layers. It starts with endothelial cell mediated transmigration of inflammatory cells from the bloodstream into the subendothelium, and then into tissue adjoining the blood vessel. Smooth muscle cells and a variety of adventitial cells reside in tunica media and tunica externa, respectively. The latter cells are a mixture of progenitor/stem cells, fibroblasts, myofibroblasts, pericytes, macrophages, and dendritic cells and respond to endothelial injury by transdifferentiation as they travel into the two inner layers, intima and media for corrective mission in the ECM composition. This chapter addresses the role of various vascular cell types and ECM components synthesized by them in maintenance of normal structure and in their contribution to major pathological processes, such as atherosclerosis, organ fibrosis, and diabetic retinopathy.
Collapse
Affiliation(s)
- Jaroslava Halper
- College of Veterinary Medicine and AU/UGA Medical Partnership, The University of Georgia, Athens, GA, United States.
| |
Collapse
|