1
|
Silva AI, Socodato R, Pinto C, Terceiro AF, Canedo T, Relvas JB, Saraiva M, Summavielle T. IL-10 and Cdc42 modulate astrocyte-mediated microglia activation in methamphetamine-induced neuroinflammation. Glia 2024; 72:1501-1517. [PMID: 38780232 DOI: 10.1002/glia.24542] [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: 01/19/2024] [Revised: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024]
Abstract
Methamphetamine (Meth) use is known to induce complex neuroinflammatory responses, particularly involving astrocytes and microglia. Building upon our previous research, which demonstrated that Meth stimulates astrocytes to release tumor necrosis factor (TNF) and glutamate, leading to microglial activation, this study investigates the role of the anti-inflammatory cytokine interleukin-10 (IL-10) in this process. Our findings reveal that the presence of recombinant IL-10 (rIL-10) counteracts Meth-induced excessive glutamate release in astrocyte cultures, which significantly reduces microglial activation. This reduction is associated with the modulation of astrocytic intracellular calcium (Ca2+) dynamics, particularly by restricting the release of Ca2+ from the endoplasmic reticulum to the cytoplasm. Furthermore, we identify the small Rho GTPase Cdc42 as a crucial intermediary in the astrocyte-to-microglia communication pathway under Meth exposure. By employing a transgenic mouse model that overexpresses IL-10 (pMT-10), we also demonstrate in vivo that IL-10 prevents Meth-induced neuroinflammation. These findings not only enhance our understanding of Meth-related neuroinflammatory mechanisms, but also suggest IL-10 and Cdc42 as putative therapeutic targets for treating Meth-induced neuroinflammation.
Collapse
Affiliation(s)
- Ana Isabel Silva
- Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Renato Socodato
- Glia Cell Biology Group, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
| | - Carolina Pinto
- Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Ana Filipa Terceiro
- Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Teresa Canedo
- Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - João Bettencourt Relvas
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Glia Cell Biology Group, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Margarida Saraiva
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Immune Regulation Group, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
| | - Teresa Summavielle
- Addiction Biology Group, i3S-Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ESS.PP, Escola Superior de Saúde do Politécnico do Porto, Porto, Portugal
| |
Collapse
|
2
|
Wan Y, Zhou J, Zhang P, Lin X, Li H. Inhibition of spinal Rac1 attenuates chronic inflammatory pain by regulating the activation of astrocytes. Cell Signal 2024; 114:110972. [PMID: 37984604 DOI: 10.1016/j.cellsig.2023.110972] [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: 08/16/2023] [Revised: 10/20/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Spinal astrocyte-mediated neuroinflammation is an important mechanism for the maintenance of chronic inflammatory pain. Previous studies have investigated that Ras-related C3 botulinum toxin substrate 1 (Rac1) is closely related to astrocyte activation after central nervous system injury. However, the role of Rac1 in astrocyte activation in chronic inflammatory pain has not been reported. METHODS Complete Freund's adjuvant (CFA)-induced chronic inflammatory pain model and LPS-stimulated astrocytes were used to investigate the role of Rac1 in astrocyte activation and the underlying mechanism. Rac1-interfering adeno-associated virus (AAV) targeting astrocytes was delivered to spinal astrocytes by intrathecal administration and a Rac1 specific inhibitor, NSC23766, was used to block cultured astrocytes. The glial fibrillary acidic protein (GFAP), proinflammatory cytokines, p-NF-κB, and nod-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome were detected by RT-qPCR, Western blotting, and immunofluorescence to investigate the activation of astrocytes. RESULTS CFA induced spinal astrocyte activation and increased the expression of active Rac1 in spinal astrocytes. Knockdown of astrocyte Rac1 alleviated chronic inflammatory pain and inhibited astrocyte activation. Inhibition of Rac1 activation in cultured astrocytes decreased the expression of GFAP and proinflammatory cytokines. Knockdown of Rac1 inhibited the increase of expression of NLRP3 inflammasome and phosphorylation of NF-κB in the spinal lumbar enlargement after CFA injection. Similarly, the inhibition of Rac1 suppressed the increase of NLRP3 inflammasome and p-NF-κB protein level after LPS stimulation. CONCLUSION Knockdown of astrocyte Rac1 attenuated CFA-induced hyperalgesia and astrocyte activation possibly by blocking the expression of NLRP3 inflammasome and phosphorylation of NF-κB.
Collapse
Affiliation(s)
- Yantong Wan
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Jieshu Zhou
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Panpan Zhang
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China
| | - Xuemei Lin
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China.
| | - Hao Li
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, China.
| |
Collapse
|
3
|
Kim C, Chen J, Ceresa BP. Chronic arsenic increases cell migration in BEAS-2B cells by increasing cell speed, cell persistence, and cell protrusion length. Exp Cell Res 2021; 408:112852. [PMID: 34599931 DOI: 10.1016/j.yexcr.2021.112852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 10/20/2022]
Abstract
There is a strong association between arsenic exposure and lung cancer development, however, the mechanism by which arsenic exposure leads to carcinogenesis is not clear. In our previous study, we observed that when BEAS-2B cells are chronically exposed to arsenic, there is an increase in secreted TGFα, as well as an increase in EGFR expression and activity. Further, these changes were broadly accompanied with an increase in cell migration. The overarching goal of this study was to acquire finer resolution of the arsenic-dependent changes in cell migration, as well as to understand the role of increased EGFR expression and activity levels in the underlying mechanisms of cell migration. To do this, we used a combination of biochemical and single cell assays, and observed chronic arsenic treatment enhancing cell migration by increasing cell speed, cell persistence and cell protrusion length. All three parameters were further increased by the addition of TGFα, indicating EGFR activity is sufficient to enhance those aspects of cell migration. In contrast, EGFR activity was necessary for the increase in cell speed, as it was reversed with an EGFR inhibitor, AG1478, but was not necessary to enhance persistence and protrusion length. From these data, we were able to isolate both EGFR-dependent and -independent features of cell migration that were enhanced by chronic arsenic exposure.
Collapse
Affiliation(s)
- Christine Kim
- Department of Pharmacology and Toxicology, University of Louisville, USA
| | - Joseph Chen
- Department of Pharmacology and Toxicology, University of Louisville, USA; Department of Bioengineering, University of Louisville, USA
| | - Brian P Ceresa
- Department of Pharmacology and Toxicology, University of Louisville, USA.
| |
Collapse
|
4
|
Xu Q, Chen J, Peng M, Duan S, Hu Y, Guo D, Geng J, Zhou J. POTEE promotes colorectal carcinoma progression via activating the Rac1/Cdc42 pathway. Exp Cell Res 2020; 390:111933. [PMID: 32142855 DOI: 10.1016/j.yexcr.2020.111933] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/22/2020] [Accepted: 02/27/2020] [Indexed: 12/21/2022]
Abstract
Current studies have shown that POTE ankyrin domain family members have high expressions as tumor antigens in malignant tumors, such as prostate cancer, ovarian cancer, breast cancer and the like. POTEE is a member of the POTE anchor protein family E. However, its role in colorectal carcinoma (CRC) has not been studied. In this study, the function of POTEE in CRC was examined for the first time and its correlation with CRC cell biological behaviors was analyzed. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR), western blotting, and immunohistochemistry revealed that POTEE was remarkably overexpressed in CRC and associated with an aggressive phenotype. We also found that POTEE was localized in the cytoplasm. In addition, downregulation of POTEE expression can notably inhibit the proliferation, migration, and invasion of CRC cell in vitro, and repressed tumor growth and metastasis in vivo. In contrast, overexpression of POTEE could promote the aggressive behaviors of CRC cells. Mechanistically, POTEE promoted CRC migration, invasion and epithelial-mesenchymal transition (EMT) by increasing the activation of Rac1 and Cdc42. To summarize, these results suggested that POTEE might serve as an oncogene for CRC tumorigenesis and progression, and may become a novel molecular marker for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Qiong Xu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jianxiong Chen
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Man Peng
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shiyu Duan
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yukun Hu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Dan Guo
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jian Geng
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Jun Zhou
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
5
|
Tam LM, Huang M, Wang Y. Targeted Quantitative Proteomics Revealed Arsenite-induced Proteasomal Degradation of RhoB in Fibroblast Cells. Chem Res Toxicol 2019; 32:1343-1350. [PMID: 31140275 PMCID: PMC6952175 DOI: 10.1021/acs.chemrestox.9b00155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Arsenic is a toxicant widely present in the environment. Previous epidemiological and animal studies support that arsenic exposure is associated with elevated incidences of lung and skin cancers. Therefore, it is important to understand the molecular mechanisms through which arsenite initiates malignant transformation of lung and skin tissues. Ras superfamily of small GTPases assumes a crucial role in many cellular processes including transcription, protein synthesis, and trafficking. In addition, small GTPase signaling is known to be altered in many types of cancer. By employing a multiple-reaction monitoring (MRM)-based targeted proteomic method, we found that the protein level of RhoB was substantially decreased in IMR90 human lung fibroblast cells upon a 12-h exposure to 5 μM NaAsO2. In addition, the protein level of ectopically expressed RhoB was found to decline in a dose-dependent manner upon arsenite exposure in HEK293T, HeLa, and GM00637 cells as well as that of endogenous RhoB protein in IMR90 cells. Moreover, the arsenite-elicited down-regulation of RhoB was found to arise from enhanced proteasomal degradation. Taken together, we demonstrated, for the first time, that exposure to arsenite could attenuate the protein expression of RhoB through proteasomal degradation.
Collapse
Affiliation(s)
- Lok Ming Tam
- Environmental Toxicology Graduate Program, University of California, Riverside, California 92521, United States
| | - Ming Huang
- Environmental Toxicology Graduate Program, University of California, Riverside, California 92521, United States
| | - Yinsheng Wang
- Environmental Toxicology Graduate Program, University of California, Riverside, California 92521, United States
- Department of Chemistry, University of California, Riverside, California 92521, United States
| |
Collapse
|
6
|
Prieto-Dominguez N, Parnell C, Teng Y. Drugging the Small GTPase Pathways in Cancer Treatment: Promises and Challenges. Cells 2019; 8:E255. [PMID: 30884855 PMCID: PMC6468615 DOI: 10.3390/cells8030255] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/08/2019] [Accepted: 03/13/2019] [Indexed: 02/07/2023] Open
Abstract
Small GTPases are a family of low molecular weight GTP-hydrolyzing enzymes that cycle between an inactive state when bound to GDP and an active state when associated to GTP. Small GTPases regulate key cellular processes (e.g., cell differentiation, proliferation, and motility) as well as subcellular events (e.g., vesicle trafficking), making them key participants in a great array of pathophysiological processes. Indeed, the dysfunction and deregulation of certain small GTPases, such as the members of the Ras and Arf subfamilies, have been related with the promotion and progression of cancer. Therefore, the development of inhibitors that target dysfunctional small GTPases could represent a potential therapeutic strategy for cancer treatment. This review covers the basic biochemical mechanisms and the diverse functions of small GTPases in cancer. We also discuss the strategies and challenges of inhibiting the activity of these enzymes and delve into new approaches that offer opportunities to target them in cancer therapy.
Collapse
Affiliation(s)
- Néstor Prieto-Dominguez
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Institute of Biomedicine (IBIOMED), University of León, León 24010, Spain.
| | | | - Yong Teng
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Department of Medical laboratory, Imaging and Radiologic Sciences, College of Allied Health, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
7
|
Proteomics analysis of Schwann cell-derived exosomes: a novel therapeutic strategy for central nervous system injury. Mol Cell Biochem 2019; 457:51-59. [PMID: 30830528 PMCID: PMC6548868 DOI: 10.1007/s11010-019-03511-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 02/14/2019] [Indexed: 12/16/2022]
Abstract
Exosomes are nanometer-sized vesicles involved in intercellular communication, and they are released by various cell types. To learn about exosomes produced by Schwann cells (SCs) and to explore their potential function in repairing the central nervous system (CNS), we isolated exosomes from supernatants of SCs by ultracentrifugation, characterized them by electron microscopy and immunoblotting and determined their protein profile using proteomic analysis. The results demonstrated that Schwann cell-derived exosomes (SCDEs) were, on average, 106.5 nm in diameter, round, and had cup-like concavity and expressed exosome markers CD9 and Alix but not tumor susceptibility gene (TSG) 101. We identified a total of 433 proteins, among which 398 proteins overlapped with the ExoCarta database. According to their specific functions, we identified 12 proteins that are closely related to CNS repair and classified them by different potential mechanisms, such as axon regeneration and inflammation inhibition. Gene Oncology analysis indicated that SCDEs are mainly involved in signal transduction and cell communication. Biological pathway analysis showed that pathways are mostly involved in exosome biogenesis, formation, uptake and axon regeneration. Among the pathways, the neurotrophin, PI3K-Akt and cAMP signaling pathways played important roles in CNS repair. Our study isolated SCDEs, unveiled their contents, presented potential neurorestorative proteins and pathways and provided a rich proteomics data resource that will be valuable for future studies of the functions of individual proteins in neurodegenerative diseases.
Collapse
|
8
|
Htike NTT, Maekawa F, Soutome H, Sano K, Maejima S, Aung KH, Tokuda M, Tsukahara S. Arsenic Exposure Induces Unscheduled Mitotic S Phase Entry Coupled with Cell Death in Mouse Cortical Astrocytes. Front Neurosci 2016; 10:297. [PMID: 27445668 PMCID: PMC4926759 DOI: 10.3389/fnins.2016.00297] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 06/13/2016] [Indexed: 11/16/2022] Open
Abstract
There is serious concern about arsenic in the natural environment, which exhibits neurotoxicity and increases the risk of neurodevelopmental disorders. Adverse effects of arsenic have been demonstrated in neurons, but it is not fully understood how arsenic affects other cell types in the brain. In the current study, we examined whether sodium arsenite (NaAsO2) affects the cell cycle, viability, and apoptosis of in vitro-cultured astrocytes isolated from the cerebral cortex of mice. Cultured astrocytes from transgenic mice expressing fluorescent ubiquitination-based cell cycle indicator (Fucci) were subjected to live imaging analysis to assess the effects of NaAsO2 (0, 1, 2, and 4 μM) on the cell cycle and number of cells. Fucci was designed to express monomeric Kusabira Orange2 (mKO2) fused with the ubiquitylation domain of hCdt1, a marker of G1 phase, and monomeric Azami Green (mAG) fused with the ubiquitylation domain of hGem, a marker of S, G2, and M phases. NaAsO2 concentration-dependently decreased the peak levels of the mAG/mKO2 emission ratio when the ratio had reached a peak in astrocytes without NaAsO2 exposure, which was due to attenuating the increase in the mAG-expressing cell number. In contrast, the mAG/mKO2 emission ratio and number of mAG-expressing cells were concentration-dependently increased by NaAsO2 before their peak levels, indicating unscheduled S phase entry. We further examined the fate of cells forced to enter S phase by NaAsO2. We found that most of these cells died up to the end of live imaging. In addition, quantification of the copy number of the glial fibrillary acidic protein gene expressed specifically in astrocytes revealed a concentration-dependent decrease caused by NaAsO2. However, NaAsO2 did not increase the amount of nucleosomes generated from DNA fragmentation and failed to alter the gene expression of molecules relevant to unscheduled S phase entry-coupled apoptosis (p21, p53, E2F1, E2F4, and Gm36566). These findings suggest that NaAsO2 adversely affects the cell cycle and viability of astrocytes by inducing unscheduled S phase entry coupled with cell death that may be caused by mechanisms other than apoptosis.
Collapse
Affiliation(s)
- Nang T T Htike
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University Saitama, Japan
| | - Fumihiko Maekawa
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies Tsukuba, Japan
| | - Haruka Soutome
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University Saitama, Japan
| | - Kazuhiro Sano
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies Tsukuba, Japan
| | - Sho Maejima
- Area of Life-NanoBio, Division of Strategy Research, Graduate School of Science and Engineering, Saitama University Saitama, Japan
| | - Kyaw H Aung
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University Saitama, Japan
| | - Masaaki Tokuda
- Department of Cell Physiology, Faculty of Medicine/Graduate School of Medicine, Kagawa University Kagawa, Japan
| | - Shinji Tsukahara
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama UniversitySaitama, Japan; Area of Life-NanoBio, Division of Strategy Research, Graduate School of Science and Engineering, Saitama UniversitySaitama, Japan
| |
Collapse
|