1
|
Keshri PK, Singh SP. Unraveling the AKT/ERK cascade and its role in Parkinson disease. Arch Toxicol 2024; 98:3169-3190. [PMID: 39136731 DOI: 10.1007/s00204-024-03829-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/25/2024] [Indexed: 09/17/2024]
Abstract
Parkinson disease represents a significant and growing burden on global healthcare systems, necessitating a deeper understanding of their underlying molecular mechanisms for the development of effective treatments. The AKT and ERK pathways play crucial roles in the disease, influencing multiple cellular pathways that support neuronal survival. Researchers have made notable progress in uncovering how these pathways are controlled by upstream kinases and how their downstream effects contribute to cell signalling. However, as we delve deeper into their intricacies, we encounter increasing complexity, compounded by the convergence of multiple signalling pathways. Many of their targets overlap with those of other kinases, and they not only affect specific substrates but also influence entire signalling networks. This review explores the intricate interplay of the AKT/ERK pathways with several other signalling cascades, including oxidative stress, endoplasmic reticulum stress, calcium homeostasis, inflammation, and autophagy, in the context of Parkinson disease. We discuss how dysregulation of these pathways contributes to disease progression and neuronal dysfunction, highlighting potential therapeutic targets for intervention. By elucidating the complex network of interactions between the AKT/ERK pathways and other signalling cascades, this review aims to provide insights into the pathogenesis of Parkinson disease and describe the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Priyanka Kumari Keshri
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Surya Pratap Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
2
|
Li Q, Li H, Zhu R, Cho WCS, Yao X, Leung FP, Tse G, Leung LK, Wong WT. TRPV2 calcium channel promotes breast cancer progression potential by activating autophagy. Cancer Cell Int 2024; 24:324. [PMID: 39334351 PMCID: PMC11438410 DOI: 10.1186/s12935-024-03506-y] [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: 07/12/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Breast cancer, the most prevalent and aggressive tumor affecting women, requires identification of disease determinants to facilitate the development of effective therapeutic strategies. Transient receptor potential vanilloid 2 (TRPV2), an ion channel highly permeable for calcium (Ca2+), is implicated in physiological and pathological processes. Nevertheless, the role of TRPV2 in breast cancer remains poorly elucidated. In this study, we found high levels of TRPV2 expression associated with advanced malignancy, thereby suggesting its potential as a biomarker for breast cancer staging. We demonstrated that TRPV2 activation promotes breast cancer cell proliferation, migration, and invasion, while silencing of TRPV2 suppresses breast cancer progression, highlighting the oncogenic role of TRPV2. Moreover, we reveal that TRPV2 facilitates cancer progression by modulating the CaMKKβ/AMPK/ULK1-autophagic axis through mediating calcium influx, providing new insights into TRPV2 as a novel therapeutic target for breast cancer treatment.
Collapse
Affiliation(s)
- Qing Li
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Huixian Li
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, 999077, China
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Ruiwen Zhu
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, 999077, China
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - William Chi Shing Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong SAR, 999077, China
| | - Xiaoqiang Yao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Fung Ping Leung
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, 999077, China
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Gary Tse
- School of Nursing and Health Studies, Hong Kong Metropolitan University, Hong Kong, 999077, China
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Lai Kwok Leung
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, 999077, China.
| | - Wing Tak Wong
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, 999077, China.
- State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong, 999077, China.
| |
Collapse
|
3
|
Yuan Y, Zhang Q, Qiu F, Kang N, Zhang Q. Targeting TRPs in autophagy regulation and human diseases. Eur J Pharmacol 2024; 977:176681. [PMID: 38821165 DOI: 10.1016/j.ejphar.2024.176681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/06/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
Transient receptor potential channels (TRPs) are widely recognized as a group of ion channels involved in various sensory perceptions, such as temperature, taste, pressure, and vision. While macroautophagy (hereafter referred to as autophagy) is primarily regulated by core machinery, the ion exchange mediated by TRPs between intracellular and extracellular compartments, as well as within organelles and the cytoplasm, plays a crucial role in autophagy regulation as an important signaling transduction mechanism. Moreover, certain TRPs can directly interact with autophagy regulatory proteins to participate in autophagy regulation. In this article, we provide an in-depth review of the current understanding of the regulatory mechanisms of autophagy, with a specific focus on TRPs. Furthermore, we highlight the potential prospects for drug development targeting TRPs in autophagy for the treatment of human diseases.
Collapse
Affiliation(s)
- Yongkang Yuan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Qiuju Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Feng Qiu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China; School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.
| | - Ning Kang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.
| | - Qiang Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.
| |
Collapse
|
4
|
Zhang J, Han H, Liu Y, Xu J, Zhang D, Wang W, Gao Y, Li Z, Qin Y. SKF96365 Inhibits Tumor Proliferation by Inducing Apoptosis and Autophagy in Human Esophageal Squamous Cell Carcinoma. Int J Genomics 2024; 2024:4501154. [PMID: 39165489 PMCID: PMC11335422 DOI: 10.1155/2024/4501154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 08/22/2024] Open
Abstract
Calcium channel blockers are emerging as a new generation of attractive anticancer drugs. SKF96365, originally thought to be a store-operated calcium entry (SOCE) inhibitor, is now often used as a TRPC channel blocker and is widely used in medical diagnostics. SKF96365 has shown antitumor effects on a variety of cancer cell lines. The objective of this study was to investigate the anticancer effect of SKF96365 on esophageal cancer in vivo and in vitro. Cell Counting Kit-8 (CCK-8) and colony formation were used to test the proliferation inhibition of SKF96365 on cell lines. Western blot and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining were used to detect cell apoptosis rates. In addition, we demonstrated the antitumor effect of SKF96365 in vivo in xenografted mice. As a result, SKF96365 significantly inhibited the proliferation of K510, K30, and EC9706 in vitro. SKF96365 induces apoptosis in three cell lines through the poly(adenosine diphosphate-ribose) polymerase (PARP), caspase-9, and BCL-2 pathways in a dose-dependent and time-dependent manner. Moreover, SKF96365 treatment also induced apoptosis and inhibited tumor growth in nude mice. The calcium channel TRPC1 was significantly downregulated by SKF96365. Autophagy was also induced during the treatment of SKF96365. In summary, SKF96365 induces apoptosis (PARP, caspase-9, and BCL-2) and autophagy (LC3-A/B) by inhibiting TRPC1 in esophageal cancer cells, thereby inhibiting tumor growth.
Collapse
Affiliation(s)
- Jiaxin Zhang
- Department of OncologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Huiqiong Han
- Department of OncologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yihan Liu
- Department of OncologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jiayao Xu
- Department of OncologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Daidi Zhang
- Department of OncologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Wenjia Wang
- Department of OncologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yaping Gao
- Department of OncologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck OncologyShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yanru Qin
- Department of OncologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
5
|
Song SE, Shin SK, Ju HY, Im SS, Song DK. Role of cytosolic and endoplasmic reticulum Ca 2+ in pancreatic beta-cells: pros and cons. Pflugers Arch 2024; 476:151-161. [PMID: 37940681 DOI: 10.1007/s00424-023-02872-2] [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: 09/04/2023] [Revised: 10/18/2023] [Accepted: 10/25/2023] [Indexed: 11/10/2023]
Abstract
Pancreatic beta cells utilize Ca2+ to secrete insulin in response to glucose. The glucose-dependent increase in cytosolic Ca2+ concentration ([Ca2+]C) activates a series of insulin secretory machinery in pancreatic beta cells. Therefore, the amount of insulin secreted in response to glucose is determined in a [Ca2+]C-dependent manner, at least within a moderate range. However, the demand for insulin secretion may surpass the capability of beta cells. Abnormal elevation of [Ca2+]C levels beyond the beta-cell endurance capacity can damage them by inducing endoplasmic reticulum (ER) stress and cell death programs such as apoptosis. Therefore, while Ca2+ is essential for the insulin secretory functions of beta cells, it could affect their survival at pathologically higher levels. Because an increase in beta-cell [Ca2+]C is inevitable under certain hazardous conditions, understanding the regulatory mechanism for [Ca2+]C is important. Therefore, this review discusses beta-cell function, survival, ER stress, and apoptosis associated with intracellular and ER Ca2+ homeostasis.
Collapse
Affiliation(s)
- Seung-Eun Song
- Department of Physiology & Obesity-Mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeol-Daeroro, Dalseo-Gu, Daegu, 42601, South Korea
| | - Su-Kyung Shin
- Department of Food Science and Nutrition, Kyungpook National University, Daegu, South Korea
| | - Hyeon Yeong Ju
- Department of Physiology & Obesity-Mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeol-Daeroro, Dalseo-Gu, Daegu, 42601, South Korea
| | - Seung-Soon Im
- Department of Physiology & Obesity-Mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeol-Daeroro, Dalseo-Gu, Daegu, 42601, South Korea
| | - Dae-Kyu Song
- Department of Physiology & Obesity-Mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeol-Daeroro, Dalseo-Gu, Daegu, 42601, South Korea.
| |
Collapse
|
6
|
Lai KC, Chueh FS, Ma YS, Chou YC, Chen JC, Liao CL, Huang YP, Peng SF. Phenethyl isothiocyanate and irinotecan synergistically induce cell apoptosis in colon cancer HCT 116 cells in vitro. ENVIRONMENTAL TOXICOLOGY 2024; 39:457-469. [PMID: 37792803 DOI: 10.1002/tox.23993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/29/2023] [Accepted: 09/21/2023] [Indexed: 10/06/2023]
Abstract
Irinotecan (IRI), an anticancer drug to treat colon cancer patients, causes cytotoxic effects on normal cells. Phenethyl isothiocyanate (PEITC), rich in common cruciferous plants, has anticancer activities (induction of cell apoptosis) in many human cancer cells, including colon cancer cells. However, the anticancer effects of IRI combined with PEITC on human colon cancer cells in vitro were unavailable. Herein, the aim of this study is to focus on the apoptotic effects of the combination of IRI and PEITC on human colon cancer HCT 116 cells in vitro. Propidium iodide (PI) exclusion and Annexin V/PI staining assays showed that IRI combined with PEITC decreased viable cell number and induced higher cell apoptosis than that of IRI or PEITC only in HCT 116 cells. Moreover, combined treatment induced higher levels of reactive oxygen species (ROS) and Ca2+ than that of IRI or PEITC only. Cells pre-treated with N-acetyl-l-cysteine (scavenger of ROS) and then treated with IRI, PEITC, or IRI combined with PEITC showed increased viable cell numbers than that of IRI or PEITC only. IRI combined with PEITC increased higher caspase-3, -8, and -9 activities than that of IRI or PEITC only by flow cytometer assay. IRI combined with PEITC induced higher levels of ER stress-, mitochondria-, and caspase-associated proteins than that of IRI or PEITC treatment only in HCT 116 cells. Based on these observations, PEITC potentiates IRI anticancer activity by promoting cell apoptosis in the human colon HCT 116 cells. Thus, PEITC may be a potential enhancer for IRI in humans as an anticolon cancer drug in the future.
Collapse
Affiliation(s)
- Kuang-Chi Lai
- Department of Medical Laboratory Science and Biotechnology, College of Medical Technology, Chung Hwa University of Medical Technology, Tainan, Taiwan
- Department of Surgery, China Medical University Beigang Hospital, Beigang, Yunlin, Taiwan
| | - Fu-Shin Chueh
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung, Taiwan
| | - Yi-Shih Ma
- School of Chinese Medicine for Post-Baccalaureate, College of Medicine, I-Shou University, Kaohsiung, Taiwan
- Department of Chinese Medicine, E-Da Cancer Hospital, Kaohsiung, Taiwan
| | - Yu-Cheng Chou
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jaw-Chyun Chen
- Department of Medicinal Botanicals and Foods on Health Applications, Da-Yeh University, Changhua, Taiwan
| | - Ching-Lung Liao
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Yi-Ping Huang
- Department of Physiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Shu-Fen Peng
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| |
Collapse
|
7
|
Nascimento Da Conceicao V, Sun Y, Chai X, Ambrus JL, Mishra BB, Singh BB. Metformin-induced activation of Ca 2+ signaling prevents immune infiltration/pathology in Sjogren's syndrome-prone mouse models. J Transl Autoimmun 2023; 7:100210. [PMID: 37711153 PMCID: PMC10497794 DOI: 10.1016/j.jtauto.2023.100210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/14/2023] [Accepted: 08/29/2023] [Indexed: 09/16/2023] Open
Abstract
Immune cell infiltration and glandular dysfunction are the hallmarks of autoimmune diseases such as primary Sjogren's syndrome (pSS), however, the mechanism(s) is unknown. Our data show that metformin-treatment induces Ca2+ signaling that restores saliva secretion and prevents immune cell infiltration in the salivary glands of IL14α-transgenic mice (IL14α), which is a model for pSS. Mechanistically, we show that loss of Ca2+ signaling is a major contributing factor, which is restored by metformin treatment, in IL14α mice. Furthermore, the loss of Ca2+ signaling leads to ER stress in salivary glands. Finally, restoration of metformin-induced Ca2+ signaling inhibited the release of alarmins and prevented the activation of ER stress that was essential for immune cell infiltration. These results suggest that loss of metformin-mediated activation of Ca2+ signaling prevents ER stress, which inhibited the release of alarmins that induces immune cell infiltration leading to salivary gland dysfunction observed in pSS.
Collapse
Affiliation(s)
- Viviane Nascimento Da Conceicao
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Yuyang Sun
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Xiufang Chai
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Julian L. Ambrus
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, School of Medicine and Biomedical Sciences, State University of New York, Buffalo, NY 14203, USA
| | - Bibhuti B. Mishra
- Department of Developmental Dentistry, School of Dentistry, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Brij B. Singh
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
8
|
Wang Y, Zhao Y, Ling Z, Luan D, Kang J, Dong X, Quan F. HD-sEVs in bovine follicular fluid regulate granulosa cell apoptosis and estradiol secretion through the autophagy pathway. Theriogenology 2023; 212:91-103. [PMID: 37717519 DOI: 10.1016/j.theriogenology.2023.09.005] [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: 01/27/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/19/2023]
Abstract
Follicular fluid (FF) is rich in extracellular vesicles (EVs), which have regulatory effects on follicular growth and oocyte development. EVs can be divided into two subtypes, i.e. HD-sEVs and LD-sEVs. In this study, HD-sEVs were successfully isolated from bovine follicular fluid (BFF) by density gradient ultracentrifugation. By western blot, quantitative polymerase chain reaction (qPCR), flow cytometry, transmission electron microscopy (TEM) and enzyme-linked immunosorbent assay (ELISA), this study found HD-sEVs promoted autophagy in bGCs by increasing the protein and mRNA expression of LC3II/LC3I ratio and Beclin1, and inhibiting the protein and mRNA expression of p62. HD-sEVs promoted mitophagy in bGCs by increasing the protein and mRNA expression of VDAC1, CTSD, and HSP60. Flow cytometry showed that HD-sEVs inhibited bGCs apoptosis rate. HD-sEVs promoted estradiol secretion by increasing steroidogenesis-associated proteins and mRNA, such as CYP19A, HSD3B in bGCs. HD-sEVs promoted autophagosome formation and mitochondrial structure swelling in bGCs, and decreased p-mTOR/mTOR ratio. The above phenomenon was reversed when wortmannin was added. Collectively, BFF HD-sEVs promote bGCs autophagy and mitophagy, inhibit bGCs apoptosis and promote estradiol secretion through the autophagy pathway-mTOR signaling pathway.
Collapse
Affiliation(s)
- Ying Wang
- Chongqing Key Laboratory of Forage &Herbivore, College of Veterinary Medicine, Southwest University, Beibei, Chongqing, 400715, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yunqi Zhao
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Zimeng Ling
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Deji Luan
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jian Kang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xiangcheng Dong
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Fusheng Quan
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
9
|
Zhu MY, Wang T, Wang HD, Wang HZ, Chen HY, Zhang S, Guo YJ, Li H, Hui H. LW-213 induces immunogenic tumor cell death via ER stress mediated by lysosomal TRPML1. Cancer Lett 2023; 577:216435. [PMID: 37806516 DOI: 10.1016/j.canlet.2023.216435] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/24/2023] [Accepted: 10/04/2023] [Indexed: 10/10/2023]
Abstract
Dying tumor cells release biological signals that exhibit antigenicity, activate cytotoxic T lymphocytes, and induce immunogenic cell death (ICD), playing a key role in immune surveillance. We demonstrate that the flavonoid LW-213 activates endoplasmic reticulum stress (ERS) in different tumor cells and that the lysosomal calcium channel TRPML1 mediates the ERS process in human cellular lymphoma Hut-102 cells. Apoptotic tumor cells induced by ERS often possess immunogenicity. Tumor cells treated with LW-213 exhibit damage-associated molecular patterns (DAMPs), including calreticulin translocation to the plasma membrane and extracellular release of ATP and HMGB1. When co-cultured with antigen-presenting cells (APCs), LW-213-treated tumor cells activated APCs. Two groups of C57BL/6J mice were inoculated with Lewis cells: a "vaccine group", which demonstrated that LW-213-treated tumor cells promote the maturation of dendritic cells and increase CD8+ T cells infiltration in the tumor microenvironment and a "pharmacodynamic group", treated with a combination of LW-213 and PD1/PD-L1 inhibitor (BMS-1), which reduced tumor growth and significantly prolonged the survival time of mice in the "pharmacodynamic group". Therefore, LW-213 can be developed as a novel ICD inducer, providing a new concept for antitumor immunotherapy.
Collapse
Affiliation(s)
- Meng-Yuan Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Ting Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Hai-di Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Hong-Zheng Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Hong-Yu Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Shuai Zhang
- The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Yong-Jian Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Hui Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China.
| | - Hui Hui
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, People's Republic of China.
| |
Collapse
|
10
|
Song SE, Shin SK, Kim YW, Do YR, Lim AK, Bae JH, Jeong GS, Im SS, Song DK. Lupenone attenuates thapsigargin-induced endoplasmic reticulum stress and apoptosis in pancreatic beta cells possibly through inhibition of protein tyrosine kinase 2 activity. Life Sci 2023; 332:122107. [PMID: 37739164 DOI: 10.1016/j.lfs.2023.122107] [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: 05/23/2023] [Revised: 09/14/2023] [Accepted: 09/17/2023] [Indexed: 09/24/2023]
Abstract
AIMS Prolonged high levels of cytokines, glucose, or free fatty acids are associated with diabetes, elevation of cytosolic Ca2+ concentration ([Ca2+]C), and depletion of Ca2+ concentration in the endoplasmic reticulum (ER) of pancreatic beta cells. This Ca2+ imbalance induces ER stress and apoptosis. Lupenone, a lupan-type triterpenoid, is beneficial in diabetes; however, its mechanism of action is yet to be clarified. This study evaluated the protective mechanism of lupenone against thapsigargin-induced ER stress and apoptosis in pancreatic beta cells. MATERIALS AND METHODS MIN6, INS-1, and native mouse islet cells were used. Western blot for protein expressions, measurement of [Ca2+]C, and in vivo glucose tolerance test were mainly performed. KEY FINDINGS Thapsigargin increased the protein levels of cleaved caspase 3, cleaved PARP, and the phosphorylated form of JNK, ATF4, and CHOP. Thapsigargin increased the interaction between stromal interaction molecule1 (Stim1) and Orai1, enhancing store-operated calcium entry (SOCE). SOCE is further activated by protein tyrosine kinase 2 (Pyk2), which is Ca2+-dependent and phosphorylates the tyrosine residue at Y361 in Stim1. Lupenone inhibited thapsigargin-mediated Pyk2 activation, suppressed [Ca2+]C, ER stress, and apoptosis. Lupenone restored impaired glucose-stimulated insulin secretion effectuated by thapsigargin and glucose intolerance in a low-dose streptozotocin-induced diabetic mouse model. SIGNIFICANCE These results suggested that lupenone attenuated thapsigargin-induced ER stress and apoptosis by inhibiting SOCE; this may be due to the hindrance of Pyk2-mediated Stim1 tyrosine phosphorylation. In beta cells that are inevitably exposed to frequent [Ca2+]C elevation, the attenuation of abnormally high SOCE would be beneficial for their survival.
Collapse
Affiliation(s)
- Seung-Eun Song
- Department of Physiology & Obesity-mediated Disease Research Center, Keimyung University School of Medicine, Daegu, South Korea
| | - Su-Kyung Shin
- Department of Food Science and Nutrition, Kyungpook National University, Daegu, South Korea
| | - Yong-Woon Kim
- Department of Physiology, Yeungnam University School of Medicine, Daegu, South Korea
| | - Young Rok Do
- Department of Internal Medicine, Keimyung University Dongsan Medical Center, Daegu, South Korea
| | - Ae Kyoung Lim
- Department of Physiology & Obesity-mediated Disease Research Center, Keimyung University School of Medicine, Daegu, South Korea
| | - Jae-Hoon Bae
- Department of Physiology & Obesity-mediated Disease Research Center, Keimyung University School of Medicine, Daegu, South Korea
| | - Gil-Saeng Jeong
- Keimyung University, College of Pharmacy, Daegu, South Korea
| | - Seung-Soon Im
- Department of Physiology & Obesity-mediated Disease Research Center, Keimyung University School of Medicine, Daegu, South Korea
| | - Dae-Kyu Song
- Department of Physiology & Obesity-mediated Disease Research Center, Keimyung University School of Medicine, Daegu, South Korea.
| |
Collapse
|
11
|
Wang Y, Zhang W, Xu G, Shi C, Wang X, Qu J, Wang H, Liu C. The role of TRPV4 in the regulation of retinal ganglion cells apoptosis in rat and mouse. Heliyon 2023; 9:e17583. [PMID: 37456002 PMCID: PMC10338314 DOI: 10.1016/j.heliyon.2023.e17583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
Retinal ganglion cell (RGC) damages are common in glaucoma, causing atrophy of the optic papilla, visual field damage, and visual loss. Transient receptor potential vanilloid 4 (TRPV4) is significantly expressed in the eyeball and is sensitive to mechanical and osmotic pressure. However, the specific role and mechanism of TRPV4 in glaucoma and RGC progression remain unclear. TRPV4 expression was detected in RGCs under different pressure culture conditions. We also explored the pressure effect on TRPV4 expression and the role and mechanism behind the functional regulation of RGCs. Immunofluorescence staining, western blotting, and TUNEL were utilized in this study. Our results established that TRPV4 was expressed in RGCs. TRPV4 expression was decreased at 40 mmHg and 60 mmHg, and the expression of BAX at 40 mmHg, 60 mmHg. Additionally, the expression of caspase 9 protein increased at 40 mmHg with the pressure increase compared with the conventional culture group. TUNEL staining revealed that the apoptosis rate of RGCs was elevated at 40 mmHg and 60 mmHg, compared with the traditional culture group. Therefore, the expression of BAX and caspase 9 increased, along with the apoptosis rate of RGCs compared with the control group. However, after TRPV4 antagonist treatment, the expression of BAX and caspase 9 decreased, and the apoptosis rate of RGCs decreased. Thus, TRPV4 may affect the mitochondrial apoptosis pathway, such as BAX and caspase 9, leading to the apoptosis of RGCs. The antagonists of TRPV4 could provide a new idea for clinically treating acute glaucoma.
Collapse
Affiliation(s)
- Yi Wang
- Department of Ophthalmology, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, 271000, China
| | - Wei Zhang
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Guozheng Xu
- Department of Physiology and Neurobiology, Functional Laboratory, Shandong First Medical University, Jinan, Shandong, 250117, China
| | - Changwei Shi
- Department of Physiology and Neurobiology, Functional Laboratory, Shandong First Medical University, Jinan, Shandong, 250117, China
| | - Xiang Wang
- School of Clinical Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271016, China
| | - Jianfeng Qu
- Medical Engineering and Technology Research Center, Shandong First Medical University, Taian, Shandong, 271000, China
| | - Hongmei Wang
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Chunhua Liu
- Department of Physiology and Neurobiology, Functional Laboratory, Shandong First Medical University, Jinan, Shandong, 250117, China
| |
Collapse
|
12
|
Radhakrishnan A, Mukherjee T, Mahish C, Kumar PS, Goswami C, Chattopadhyay S. TRPA1 activation and Hsp90 inhibition synergistically downregulate macrophage activation and inflammatory responses in vitro. BMC Immunol 2023; 24:16. [PMID: 37391696 PMCID: PMC10314470 DOI: 10.1186/s12865-023-00549-0] [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: 09/20/2022] [Accepted: 06/14/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND Transient receptor potential ankyrin 1 (TRPA1) channels are known to be actively involved in various pathophysiological conditions, including neuronal inflammation, neuropathic pain, and various immunological responses. Heat shock protein 90 (Hsp90), a cytoplasmic molecular chaperone, is well-reported for various cellular and physiological processes. Hsp90 inhibition by various molecules has garnered importance for its therapeutic significance in the downregulation of inflammation and are proposed as anti-cancer drugs. However, the possible role of TRPA1 in the Hsp90-associated modulation of immune responses remains scanty. RESULTS Here, we have investigated the role of TRPA1 in regulating the anti-inflammatory effect of Hsp90 inhibition via 17-(allylamino)-17-demethoxygeldanamycin (17-AAG) in lipopolysaccharide (LPS) or phorbol 12-myristate 13-acetate (PMA) stimulation in RAW 264.7, a mouse macrophage cell lines and PMA differentiated THP-1, a human monocytic cell line similar to macrophages. Activation of TRPA1 with Allyl isothiocyanate (AITC) is observed to execute an anti-inflammatory role via augmenting Hsp90 inhibition-mediated anti-inflammatory responses towards LPS or PMA stimulation in macrophages, whereas inhibition of TRPA1 by 1,2,3,6-Tetrahydro-1,3-dimethyl-N-[4-(1-methylethyl)phenyl]-2,6-dioxo-7 H-purine-7-acetamide,2-(1,3-Dimethyl-2,6-dioxo-1,2,3,6-tetrahydro-7 H-purin-7-yl)-N-(4-isopropylphenyl)acetamide (HC-030031) downregulates these developments. LPS or PMA-induced macrophage activation was found to be regulated by TRPA1. The same was confirmed by studying the levels of activation markers (major histocompatibility complex II (MHCII), cluster of differentiation (CD) 80 (CD80), and CD86, pro-inflammatory cytokines (tumor necrosis factor (TNF) and interleukin 6 (IL-6)), NO (nitric oxide) production, differential expression of mitogen-activated protein kinase (MAPK) signaling pathways (p-p38 MAPK, phospho-extracellular signal-regulated kinase 1/2 (p-ERK 1/2), and phosphor-stress-activated protein kinase/c-Jun N-terminal kinase (p-SAPK/JNK)), and induction of apoptosis. Additionally, TRPA1 has been found to be an important contributor to intracellular calcium levels toward Hsp90 inhibition in LPS or PMA-stimulated macrophages. CONCLUSION This study indicates a significant role of TRPA1 in Hsp90 inhibition-mediated anti-inflammatory developments in LPS or PMA-stimulated macrophages. Activation of TRPA1 and inhibition of Hsp90 has synergistic roles towards regulating inflammatory responses associated with macrophages. The role of TRPA1 in Hsp90 inhibition-mediated modulation of macrophage responses may provide insights towards designing future novel therapeutic approaches to regulate various inflammatory responses.
Collapse
Affiliation(s)
- Anukrishna Radhakrishnan
- National Institute of Science Education and Research, an Off-campus Centre (OCC) of Homi Bhabha National Institute, Bhubaneswar, Odisha 752050 India
| | - Tathagata Mukherjee
- National Institute of Science Education and Research, an Off-campus Centre (OCC) of Homi Bhabha National Institute, Bhubaneswar, Odisha 752050 India
| | - Chandan Mahish
- National Institute of Science Education and Research, an Off-campus Centre (OCC) of Homi Bhabha National Institute, Bhubaneswar, Odisha 752050 India
| | - P Sanjai Kumar
- Institute of Life Sciences, Nalco Nagar Rd, NALCO Square, NALCO Nagar, Chandrasekharpur, Bhubaneswar, Odisha 751023 India
| | - Chandan Goswami
- National Institute of Science Education and Research, an Off-campus Centre (OCC) of Homi Bhabha National Institute, Bhubaneswar, Odisha 752050 India
| | - Subhasis Chattopadhyay
- National Institute of Science Education and Research, an Off-campus Centre (OCC) of Homi Bhabha National Institute, Bhubaneswar, Odisha 752050 India
| |
Collapse
|
13
|
Gene expression of TRPMLs and its regulation by pathogen stimulation. Gene 2023; 864:147291. [PMID: 36813061 DOI: 10.1016/j.gene.2023.147291] [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: 12/04/2022] [Revised: 01/18/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023]
Abstract
The transient receptor potential mucolipin (TRPML) subfamily in mammalian has three members, namely TRPML1, TRPML2, and TRPML3, who play key roles in regulating intracellular Ca2+ homeostasis, endosomal pH, membrane trafficking and autophagy. Previous studies had shown that three TRPMLs are closely related to the occurrence of pathogen invasion and immune regulation in some immune tissues or cells, but the relationship between TRPMLs expression and pathogen invasion in lung tissue or cell remains elusive. Here, we investigated the expression distribution of three TRPML channels in mouse different tissues by qRT-PCR, and then found that all three TRPMLs were highly expressed in the mouse lung tissue, as well as mouse spleen and kidney tissues. The expression of TRPML1 or TRPML3 in all three mouse tissues had a significant down-regulation after the treatment of Salmonella or LPS, but TRPML2 expression showed a remarkable increase. Consistently, TRPML1 or TRPML3 but not TRPML2 in A549 cells also displayed a decreased expression induced by LPS stimulation, which shared a similar regulation pattern in the mouse lung tissue. Furthermore, the treatment of the TRPML1 or TRPML3 specific activator induced a dose-dependent up-regulation of inflammatory factors IL-1β, IL-6 and TNFα, suggesting that TRPML1 and TRPML3 are likely to play an important role in immune and inflammatory regulation. Together, our study identified the gene expression of TRPMLs induced by pathogen stimulation in vivo and in vitro, which may provide novel targets for innate immunity or pathogen regulation.
Collapse
|
14
|
Wei Y, Khalaf AT, Rui C, Abdul Kadir SY, Zainol J, Oglah Z. The Emergence of TRP Channels Interactome as a Potential Therapeutic Target in Pancreatic Ductal Adenocarcinoma. Biomedicines 2023; 11:biomedicines11041164. [PMID: 37189782 DOI: 10.3390/biomedicines11041164] [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: 03/05/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Integral membrane proteins, known as Transient Receptor Potential (TRP) channels, are cellular sensors for various physical and chemical stimuli in the nervous system, respiratory airways, colon, pancreas, bladder, skin, cardiovascular system, and eyes. TRP channels with nine subfamilies are classified by sequence similarity, resulting in this superfamily's tremendous physiological functional diversity. Pancreatic Ductal Adenocarcinoma (PDAC) is the most common and aggressive form of pancreatic cancer. Moreover, the development of effective treatment methods for pancreatic cancer has been hindered by the lack of understanding of the pathogenesis, partly due to the difficulty in studying human tissue samples. However, scientific research on this topic has witnessed steady development in the past few years in understanding the molecular mechanisms that underlie TRP channel disturbance. This brief review summarizes current knowledge of the molecular role of TRP channels in the development and progression of pancreatic ductal carcinoma to identify potential therapeutic interventions.
Collapse
Affiliation(s)
- Yuanyuan Wei
- Basic Medical College, Chengdu University, Chengdu 610106, China
| | | | - Cao Rui
- Basic Medical College, Chengdu University, Chengdu 610106, China
| | - Samiah Yasmin Abdul Kadir
- Faculty of Medicine, Widad University College, BIM Point, Bandar Indera Mahkota, Kuantan 25200, Malaysia
| | - Jamaludin Zainol
- Faculty of Medicine, Widad University College, BIM Point, Bandar Indera Mahkota, Kuantan 25200, Malaysia
| | - Zahraa Oglah
- School of Science, Auckland University of Technology (AUT), 55 Wellesley Street, Auckland 1010, New Zealand
| |
Collapse
|
15
|
Vestuto V, Di Sarno V, Musella S, Di Dona G, Moltedo O, Gomez-Monterrey IM, Bertamino A, Ostacolo C, Campiglia P, Ciaglia T. New Frontiers on ER Stress Modulation: Are TRP Channels the Leading Actors? Int J Mol Sci 2022; 24:185. [PMID: 36613628 PMCID: PMC9820239 DOI: 10.3390/ijms24010185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
The endoplasmic reticulum (ER) is a dynamic structure, playing multiple roles including calcium storage, protein synthesis and lipid metabolism. During cellular stress, variations in ER homeostasis and its functioning occur. This condition is referred as ER stress and generates a cascade of signaling events termed unfolded protein response (UPR), activated as adaptative response to mitigate the ER stress condition. In this regard, calcium levels play a pivotal role in ER homeostasis and therefore in cell fate regulation since calcium signaling is implicated in a plethora of physiological processes, but also in disease conditions such as neurodegeneration, cancer and metabolic disorders. A large body of emerging evidence highlighted the functional role of TRP channels and their ability to promote cell survival or death depending on endoplasmic reticulum stress resolution, making them an attractive target. Thus, in this review we focused on the TRP channels' correlation to UPR-mediated ER stress in disease pathogenesis, providing an overview of their implication in the activation of this cellular response.
Collapse
Affiliation(s)
- Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Veronica Di Sarno
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Simona Musella
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Giorgio Di Dona
- Pineta Grande Hospital, Via Domiziana, km 30/00, 81030 Castel Volturno, CE, Italy
| | - Ornella Moltedo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | | | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Carmine Ostacolo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Napoli, NA, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
- European Biomedical Research Institute of Salerno, Via S. De Renzi 50, 84125 Salerno, SA, Italy
| | - Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| |
Collapse
|
16
|
Sun Y, Zboril EK, De La Chapa JJ, Chai X, Da Conceicao VN, Valdez MC, McHardy SF, Gonzales CB, Singh BB. Inhibition of Ca 2+ entry by capsazepine analog CIDD-99 prevents oral squamous carcinoma cell proliferation. Front Physiol 2022; 13:969000. [PMID: 36187775 PMCID: PMC9521718 DOI: 10.3389/fphys.2022.969000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Oral cancer patients have a poor prognosis, with approximately 66% of patients surviving 5-years after diagnosis. Treatments for oral cancer are limited and have many adverse side effects; thus, further studies are needed to develop drugs that are more efficacious. To achieve this objective, we developed CIDD-99, which produces cytotoxic effects in multiple oral squamous cell carcinoma (OSCC) cell lines. While we demonstrated that CIDD-99 induces ER stress and apoptosis in OSCC, the mechanism was unclear. Investigation of the Bcl-family of proteins showed that OSCC cells treated with CIDD-99 undergo downregulation of Bcl-XL and Bcl-2 anti-apoptotic proteins and upregulation of Bax (pro-apoptotic). Importantly, OSCC cells treated with CIDD-99 displayed decreased calcium signaling in a dose and time-dependent manner, suggesting that blockage of calcium signaling is the key mechanism that induces cell death in OSCC. Indeed, CIDD-99 anti-proliferative effects were reversed by the addition of exogenous calcium. Moreover, electrophysiological properties further established that calcium entry was via the non-selective TRPC1 channel and prolonged CIDD-99 incubation inhibited STIM1 expression. CIDD-99 inhibition of calcium signaling also led to ER stress and inhibited mitochondrial complexes II and V in vitro. Taken together, these findings suggest that inhibition of TRPC mediates induction of ER stress and mitochondrial dysfunction as a part of the cellular response to CIDD-99 in OSCC.
Collapse
Affiliation(s)
- Yuyang Sun
- Department of Periodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Emily K. Zboril
- Department of Periodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Jorge J. De La Chapa
- Department of Comprehensive Dentistry, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Xiufang Chai
- Department of Periodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX, United States
| | | | - Matthew C. Valdez
- Department of Chemistry and the Center for Innovative Drug Discovery, University of Texas at San Antonio, San Antonio, TX, United States
| | - Stanton F. McHardy
- Department of Chemistry and the Center for Innovative Drug Discovery, University of Texas at San Antonio, San Antonio, TX, United States
| | - Cara B. Gonzales
- Department of Comprehensive Dentistry, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Brij B. Singh
- Department of Periodontics, School of Dentistry, University of Texas Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
17
|
Collins HE, Zhang D, Chatham JC. STIM and Orai Mediated Regulation of Calcium Signaling in Age-Related Diseases. FRONTIERS IN AGING 2022; 3:876785. [PMID: 35821821 PMCID: PMC9261457 DOI: 10.3389/fragi.2022.876785] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/30/2022] [Indexed: 01/19/2023]
Abstract
Tight spatiotemporal regulation of intracellular Ca2+ plays a critical role in regulating diverse cellular functions including cell survival, metabolism, and transcription. As a result, eukaryotic cells have developed a wide variety of mechanisms for controlling Ca2+ influx and efflux across the plasma membrane as well as Ca2+ release and uptake from intracellular stores. The STIM and Orai protein families comprising of STIM1, STIM2, Orai1, Orai2, and Orai3, are evolutionarily highly conserved proteins that are core components of all mammalian Ca2+ signaling systems. STIM1 and Orai1 are considered key players in the regulation of Store Operated Calcium Entry (SOCE), where release of Ca2+ from intracellular stores such as the Endoplasmic/Sarcoplasmic reticulum (ER/SR) triggers Ca2+ influx across the plasma membrane. SOCE, which has been widely characterized in non-excitable cells, plays a central role in Ca2+-dependent transcriptional regulation. In addition to their role in Ca2+ signaling, STIM1 and Orai1 have been shown to contribute to the regulation of metabolism and mitochondrial function. STIM and Orai proteins are also subject to redox modifications, which influence their activities. Considering their ubiquitous expression, there has been increasing interest in the roles of STIM and Orai proteins in excitable cells such as neurons and myocytes. While controversy remains as to the importance of SOCE in excitable cells, STIM1 and Orai1 are essential for cellular homeostasis and their disruption is linked to various diseases associated with aging such as cardiovascular disease and neurodegeneration. The recent identification of splice variants for most STIM and Orai isoforms while complicating our understanding of their function, may also provide insight into some of the current contradictions on their roles. Therefore, the goal of this review is to describe our current understanding of the molecular regulation of STIM and Orai proteins and their roles in normal physiology and diseases of aging, with a particular focus on heart disease and neurodegeneration.
Collapse
Affiliation(s)
- Helen E. Collins
- Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Dingguo Zhang
- Division of Molecular and Cellular Pathology, Department of PathologyUniversity of Alabama at Birmingham, Birmingham, AL, United States
| | - John C. Chatham
- Division of Molecular and Cellular Pathology, Department of PathologyUniversity of Alabama at Birmingham, Birmingham, AL, United States,*Correspondence: John C. Chatham,
| |
Collapse
|
18
|
YAO Y, SHI Y, AN P, ZHANG R, WANG Z, HU X, WAN Y. Optimization of preparation of calcium propionate from eggshell by Response Surface Methodology (RSM). FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.25322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
19
|
Guan PP, Cao LL, Yang Y, Wang P. Calcium Ions Aggravate Alzheimer's Disease Through the Aberrant Activation of Neuronal Networks, Leading to Synaptic and Cognitive Deficits. Front Mol Neurosci 2021; 14:757515. [PMID: 34924952 PMCID: PMC8674839 DOI: 10.3389/fnmol.2021.757515] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/04/2021] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease that is characterized by the production and deposition of β-amyloid protein (Aβ) and hyperphosphorylated tau, leading to the formation of β-amyloid plaques (APs) and neurofibrillary tangles (NFTs). Although calcium ions (Ca2+) promote the formation of APs and NFTs, no systematic review of the mechanisms by which Ca2+ affects the development and progression of AD has been published. Therefore, the current review aimed to fill the gaps between elevated Ca2+ levels and the pathogenesis of AD. Specifically, we mainly focus on the molecular mechanisms by which Ca2+ affects the neuronal networks of neuroinflammation, neuronal injury, neurogenesis, neurotoxicity, neuroprotection, and autophagy. Furthermore, the roles of Ca2+ transporters located in the cell membrane, endoplasmic reticulum (ER), mitochondria and lysosome in mediating the effects of Ca2+ on activating neuronal networks that ultimately contribute to the development and progression of AD are discussed. Finally, the drug candidates derived from herbs used as food or seasoning in Chinese daily life are summarized to provide a theoretical basis for improving the clinical treatment of AD.
Collapse
Affiliation(s)
- Pei-Pei Guan
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Long-Long Cao
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Yi Yang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Pu Wang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| |
Collapse
|
20
|
Nascimento Da Conceicao V, Sun Y, Ramachandran K, Chauhan A, Raveendran A, Venkatesan M, DeKumar B, Maity S, Vishnu N, Kotsakis GA, Worley PF, Gill DL, Mishra BB, Madesh M, Singh BB. Resolving macrophage polarization through distinct Ca 2+ entry channel that maintains intracellular signaling and mitochondrial bioenergetics. iScience 2021; 24:103339. [PMID: 34816101 PMCID: PMC8591423 DOI: 10.1016/j.isci.2021.103339] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/30/2021] [Accepted: 10/20/2021] [Indexed: 01/21/2023] Open
Abstract
Transformation of naive macrophages into classically (M1) or alternatively (M2) activated macrophages regulates the inflammatory response. Here, we identified that distinct Ca2+ entry channels determine the IFNγ-induced M1 or IL-4-induced M2 transition. Naive or M2 macrophages exhibit a robust Ca2+ entry that was dependent on Orai1 channels, whereas the M1 phenotype showed a non-selective TRPC1 current. Blockade of Ca2+ entry suppresses pNF-κB/pJNK/STAT1 or STAT6 signaling events and consequently lowers cytokine production that is essential for M1 or M2 functions. Of importance, LPS stimulation shifted M2 cells from Orai1 toward TRPC1-mediated Ca2+ entry and TRPC1-/- mice exhibited transcriptional changes that suppress pro-inflammatory cytokines. In contrast, Orai1-/- macrophages showed a decrease in anti-inflammatory cytokines and exhibited a suppression of mitochondrial oxygen consumption rate and inhibited mitochondrial shape transition specifically in the M2 cells. Finally, alterations in TRPC1 or Orai1 expression determine macrophage polarization suggesting a distinct role of Ca2+ channels in modulating macrophage transformation.
Collapse
Affiliation(s)
| | - Yuyang Sun
- Department of Periodontics, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Karthik Ramachandran
- Department of Medicine, Cardiology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Arun Chauhan
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Amritha Raveendran
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Manigandan Venkatesan
- Department of Medicine, Cardiology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Bony DeKumar
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Soumya Maity
- Department of Medicine, Cardiology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Neelanjan Vishnu
- Department of Medicine, Cardiology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - George A. Kotsakis
- Department of Periodontics, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Paul F. Worley
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Donald L. Gill
- Department of Cellular and Molecular Physiology, Penn State Hershey College of Medicine, Hershey, PA 17033, USA
| | - Bibhuti B. Mishra
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Muniswamy Madesh
- Department of Medicine, Cardiology Division, Center for Precision Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Brij B. Singh
- Department of Periodontics, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
21
|
Bektur Aykanat NE, Şahin E, Kaçar S, Bağcı R, Karakaya Ş, Burukoğlu Dönmez D, Şahintürk V. Cardiac hypertrophy caused by hyperthyroidism in rats: the role of ATF-6 and TRPC1 channels. Can J Physiol Pharmacol 2021; 99:1226-1233. [PMID: 34283935 DOI: 10.1139/cjpp-2021-0260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hyperthyroidism influences the development of cardiac hypertrophy. Transient receptor potential canonical channels (TRPCs) and endoplasmic reticulum (ER) stress are regarded as critical pathways in cardiac hypertrophy. Hence, we aimed to identify the TRPCs associated with ER stress in hyperthyroidism-induced cardiac hypertrophy. Twenty adult Wistar albino male rats were used in the study. The control group was fed with standard food and tap water. The group with hyperthyroidism was also fed with standard rat food, along with tap water that contained 12 mg/L of thyroxine (T4) for 4 weeks. At the end of the fourth week, the serum-free triiodothyronine (T3), T4, and thyroid-stimulating hormone (TSH) levels of the groups were measured. The left ventricle of each rat was used for histochemistry, immunohistochemistry, Western blot, total antioxidant capacity (TAC), and total oxidant status (TOS) analysis. As per our results, activating transcription factor 6 (ATF-6), inositol-requiring kinase 1 (IRE-1), and TRPC1, which play a significant role in cardiac hypertrophy caused by hyperthyroidism, showed increased activation. Moreover, TOS and serum-free T3 levels increased, while TAC and TSH levels decreased. With the help of the literature review in our study, we could, for the first time, indicate that the increased activation of ATF-6, IRE-1, and TRPC1-induced deterioration of the Ca2+ ion balance leads to hypertrophy in hyperthyroidism due to heart failure.
Collapse
Affiliation(s)
| | - Erhan Şahin
- Department of Histology and Embryology, Eskişehir Osmangazi University Faculty of Medicine, Eskişehir, Turkey
| | - Sedat Kaçar
- Department of Histology and Embryology, Eskişehir Osmangazi University Faculty of Medicine, Eskişehir, Turkey
| | - Rıdvan Bağcı
- Adana City Training and Research Hospital, Adana, Turkey
| | - Şerife Karakaya
- Department of Histology and Embryology, Eskişehir Osmangazi University Faculty of Medicine, Eskişehir, Turkey
| | - Dilek Burukoğlu Dönmez
- Department of Histology and Embryology, Eskişehir Osmangazi University Faculty of Medicine, Eskişehir, Turkey
| | - Varol Şahintürk
- Department of Histology and Embryology, Eskişehir Osmangazi University Faculty of Medicine, Eskişehir, Turkey
| |
Collapse
|
22
|
Recent advances in active targeting of nanomaterials for anticancer drug delivery. Adv Colloid Interface Sci 2021; 296:102509. [PMID: 34455211 DOI: 10.1016/j.cis.2021.102509] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/24/2021] [Accepted: 08/15/2021] [Indexed: 12/15/2022]
Abstract
One of the challenges in cancer chemotherapy is the low target to non-target ratio of therapeutic agents which incur severe adverse effect on the healthy tissues. In this regard, nanomaterials have tremendous potential for impacting cancer therapy by altering the toxicity profile of the drug. Some of the striking advantages provided by the nanocarriers mediated targeted drug delivery are relatively high build-up of drug concentration at the tumor site, improved drug content in the formulation and enhanced colloidal stability. Further, nanocarriers with tumor-specific moieties can be targeted to the cancer cell through cell surface receptors, tumor antigens and tumor vasculatures with high affinity and accuracy. Moreover, it overcomes the bottleneck of aimless drug biodistribution, undesired toxicity and heavy dosage of administration. This review discusses the recent developments in active targeting of nanomaterials for anticancer drug delivery through cancer cell surface targeting, organelle specific targeting and tumor microenvironment targeting strategies. Special emphasis has been given towards cancer cell surface and organelle specific targeting as delivery of anticancer drugs through these routes have made paradigm change in cancer management. Further, the current challenges and future prospects of nanocarriers mediated active drug targeting are also demonstrated.
Collapse
|
23
|
Sharma A, Ramena GT, Elble RC. Advances in Intracellular Calcium Signaling Reveal Untapped Targets for Cancer Therapy. Biomedicines 2021; 9:1077. [PMID: 34572262 PMCID: PMC8466575 DOI: 10.3390/biomedicines9091077] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/15/2021] [Accepted: 07/18/2021] [Indexed: 02/07/2023] Open
Abstract
Intracellular Ca2+ distribution is a tightly regulated process. Numerous Ca2+ chelating, storage, and transport mechanisms are required to maintain normal cellular physiology. Ca2+-binding proteins, mainly calmodulin and calbindins, sequester free intracellular Ca2+ ions and apportion or transport them to signaling hubs needing the cations. Ca2+ channels, ATP-driven pumps, and exchangers assist the binding proteins in transferring the ions to and from appropriate cellular compartments. Some, such as the endoplasmic reticulum, mitochondria, and lysosomes, act as Ca2+ repositories. Cellular Ca2+ homeostasis is inefficient without the active contribution of these organelles. Moreover, certain key cellular processes also rely on inter-organellar Ca2+ signaling. This review attempts to encapsulate the structure, function, and regulation of major intracellular Ca2+ buffers, sensors, channels, and signaling molecules before highlighting how cancer cells manipulate them to survive and thrive. The spotlight is then shifted to the slow pace of translating such research findings into anticancer therapeutics. We use the PubMed database to highlight current clinical studies that target intracellular Ca2+ signaling. Drug repurposing and improving the delivery of small molecule therapeutics are further discussed as promising strategies for speeding therapeutic development in this area.
Collapse
Affiliation(s)
- Aarushi Sharma
- Department of Pharmacology and Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL 62702, USA;
| | - Grace T. Ramena
- Department of Aquaculture, University of Arkansas, Pine Bluff, AR 71601, USA;
| | - Randolph C. Elble
- Department of Pharmacology and Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL 62702, USA;
| |
Collapse
|
24
|
Sun Y, Sukumaran P, Singh BB. Sigma1 Receptor Inhibits TRPC1-Mediated Ca 2+ Entry That Promotes Dopaminergic Cell Death. Cell Mol Neurobiol 2021; 41:1245-1255. [PMID: 32514827 DOI: 10.1007/s10571-020-00892-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 05/28/2020] [Indexed: 12/24/2022]
Abstract
Regulation of Ca2+ homeostasis is essential for neuronal function and its survival. Recent data suggest that TRPC1 function as the endogenous store-mediated Ca2+ entry channel in dopaminergic cells, and loss of TRPC1 function leads to neurodegeneration; however, its regulation is not fully identified. Here we provide evidence that the sigma 1 receptor contributes to the loss of dopaminergic cells by blocking TRPC1-mediated Ca2+ entry. Importantly, downregulation of sigma 1 receptor expression significantly decreased neurotoxin-induced loss of dopaminergic cells as measured by MTT assays and caspase activity was also inhibited. Importantly, sigma 1 receptor inhibited TRPC1-mediated Ca2+ entry and silencing of sigma 1 receptor significantly restored store-dependent Ca2+ influx. Although co-immunoprecipitation failed to show an interaction between the TRPC1 and sigma 1 receptor, store depletion promoted a decrease in the sigma 1 receptor-STIM1 association. Neurotoxin-induced loss of Ca2+ entry was significantly restored in cells that had decreased sigma 1 receptor expression. Furthermore, TRPC1 or STIM1 silencing inhibited store-mediated Ca2+ entry, which was further increased upon the downregulation of the sigma 1 receptor expression. TRPC1 silencing prevented the increased neuroprotection and caspase activity observed upon the downregulation of sigma 1 receptor. Finally, sigma 1 receptor activation also significantly decreased TRPC1-mediated Ca2+ entry and lead to an increase in neurodegeneration. In contrast, addition of sigma 1 receptor antagonist prevented neurotoxin-induced neurodegeneration and facilitated TRPC1-mediated Ca2+ influx. Together these results suggest that the sigma 1 receptor is involved in the inhibition of TRPC1- mediated Ca2+ entry, which leads to the degeneration in the dopaminergic cells, and prevention of sigma 1 receptor function could protect neuronal cell death as observed in Parkinson's disease.
Collapse
Affiliation(s)
- Yuyang Sun
- Department of Periodontics, University of Texas Health Science Center San Antonio, San Antonio, TX, 78229, USA
| | - Pramod Sukumaran
- Department of Periodontics, University of Texas Health Science Center San Antonio, San Antonio, TX, 78229, USA
| | - Brij B Singh
- Department of Periodontics, University of Texas Health Science Center San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
25
|
BEKTUR AYKANAT NE, ŞAHİN E, KAÇAR S, BAĞCI R, KARAKAYA Ş, BURUKOĞLU DÖNMEZ D, ŞAHİNTÜRK V. Investigation of the effect of hyperthyroidism on endoplasmic reticulum stress and tran- sient receptor potential canonical 1 channel in the kidney. Turk J Med Sci 2021; 51:1554-1563. [PMID: 33754657 PMCID: PMC8283502 DOI: 10.3906/sag-2007-109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 03/20/2021] [Indexed: 11/07/2022] Open
Abstract
Background/aim Hyperthyroidism is associated with results in increased glomerular filtration rate as well as increased renin-angio- tensin-aldosterone activation. The disturbance of Ca2+ homeostasis in the endoplasmic reticulum (ER) is associated with many diseases, including diabetic nephropathy and hyperthyroidism. Transient receptor potential canonical 1 (TRPC1) channel is the first cloned TRPC family protein. Although it is expressed in many places in the kidney, its function is uncertain. TRPC1 is involved in regulating Ca2+ homeostasis, and its upregulation increases ER Ca2+ level, activates the unfolded protein response, which leads to cellular damage in the kidney. This study investigated the role of TRPC1 in the kidneys of hyperthyroid rats in terms of ER stress markers that are gluco- se-regulated protein 78 (GRP78), activating transcription factor 6 (ATF6), (protein kinase R (PKR)-like endoplasmic reticulum kinase) (PERK), Inositol-requiring enzyme 1 (IRE1). Materials and methods Twenty male rats were assigned into control and hyperthyroid groups (n = 10). Hyperthyroidism was induced by adding 12 mg/L thyroxine into the drinking water of rats for 4 weeks. The serum-free T3 and T4 (fT3, fT4), TSH, blood urea nitrogen (BUN), and creatinine levels were measured. The histochemical analysis of kidney sections for morphological changes and also im- munohistochemical and western blot analysis of kidney sections were performed for GRP78, ATF6, PERK, IRE1, TRPC1 antibodies. Results TSH, BUN, and creatinine levels decreased while fT3 and fT4 levels increased in the hyperthyroid rat. The morphologic analy- sis resulted in the capillary basal membrane thickening in glomeruli and also western blot, and immunohistochemical results showed an increase in TRPC1, GRP78, and ATF6 in the hyperthyroid rat (p < 0.05). Conclusion In conclusion, in our study, we showed for the first time that the relationship between ER stress and TRPC1, and their increased expression caused renal damage in hyperthyroid rats.
Collapse
Affiliation(s)
| | - Erhan ŞAHİN
- Department of Histology and Embryology, Faculty of Medicine, Osmangazi University, AnkaraTurkey
| | - Sedat KAÇAR
- Department of Histology and Embryology, Faculty of Medicine, Osmangazi University, AnkaraTurkey
| | - Rıdvan BAĞCI
- Department of IVF Unit Andrology Laboratory, Adana City Education and Research Hospital, AdanaTurkey
| | - Şerife KARAKAYA
- Department of Histology and Embryology, Faculty of Medicine, Osmangazi University, AnkaraTurkey
| | - Dilek BURUKOĞLU DÖNMEZ
- Department of Histology and Embryology, Faculty of Medicine, Osmangazi University, AnkaraTurkey
| | - Varol ŞAHİNTÜRK
- Department of Histology and Embryology, Faculty of Medicine, Osmangazi University, AnkaraTurkey
| |
Collapse
|
26
|
Sanjai Kumar P, Nayak TK, Mahish C, Sahoo SS, Radhakrishnan A, De S, Datey A, Sahu RP, Goswami C, Chattopadhyay S, Chattopadhyay S. Inhibition of transient receptor potential vanilloid 1 (TRPV1) channel regulates chikungunya virus infection in macrophages. Arch Virol 2020; 166:139-155. [PMID: 33125586 DOI: 10.1007/s00705-020-04852-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 09/08/2020] [Indexed: 11/29/2022]
Abstract
Chikungunya virus (CHIKV), a virus that induces pathogenic inflammatory host immune responses, is re-emerging worldwide, and there are currently no established antiviral control measures. Transient receptor potential vanilloid 1 (TRPV1), a non-selective Ca2+-permeable ion channel, has been found to regulate various host inflammatory responses including several viral infections. Immune responses to CHIKV infection in host macrophages have been reported recently. However, the possible involvement of TRPV1 during CHIKV infection in host macrophages has not been studied. Here, we investigated the possible role of TRPV1 in CHIKV infection of the macrophage cell line RAW 264.7. It was found that CHIKV infection upregulates TRPV1 expression in macrophages. To confirm this observation, the TRPV1-specific modulators 5'-iodoresiniferatoxin (5'-IRTX, a TRPV1 antagonist) and resiniferatoxin (RTX, a TRPV1 agonist) were used. Our results indicated that TRPV1 inhibition leads to a reduction in CHIKV infection, whereas TRPV1 activation significantly enhances CHIKV infection. Using a plaque assay and a time-of-addition assay, it was observed that functional modulation of TRPV1 affects the early stages of the viral lifecycle in RAW 264.7 cells. Moreover, CHIKV infection was found to induce of pNF-κB (p65) expression and nuclear localization. However, both activation and inhibition of TRPV1 were found to enhance the expression and nuclear localization of pNF-κB (p65) and production of pro-inflammatory TNF and IL-6 during CHIKV infection. In addition, it was demonstrated by Ca2+ imaging that TRPV1 regulates Ca2+ influx during CHIKV infection. Hence, the current findings highlight a potentially important regulatory role of TRPV1 during CHIKV infection in macrophages. This study might also have broad implications in the context of other viral infections as well.
Collapse
Affiliation(s)
- P Sanjai Kumar
- School of Biological Sciences, National Institute of Science Education & Research, Bhubaneswar, HBNI, Jatni, Khurda, Odisha, 752050, India
| | - Tapas K Nayak
- School of Biological Sciences, National Institute of Science Education & Research, Bhubaneswar, HBNI, Jatni, Khurda, Odisha, 752050, India.,Infectious Disease Biology, Institute of Life Sciences, (Autonomous Institute of Department of Biotechnology, Government of India), Nalco Square, Bhubaneswar, Odisha, 751023, India
| | - Chandan Mahish
- School of Biological Sciences, National Institute of Science Education & Research, Bhubaneswar, HBNI, Jatni, Khurda, Odisha, 752050, India
| | - Subhransu S Sahoo
- School of Biological Sciences, National Institute of Science Education & Research, Bhubaneswar, HBNI, Jatni, Khurda, Odisha, 752050, India
| | - Anukrishna Radhakrishnan
- School of Biological Sciences, National Institute of Science Education & Research, Bhubaneswar, HBNI, Jatni, Khurda, Odisha, 752050, India
| | - Saikat De
- Infectious Disease Biology, Institute of Life Sciences, (Autonomous Institute of Department of Biotechnology, Government of India), Nalco Square, Bhubaneswar, Odisha, 751023, India
| | - Ankita Datey
- Infectious Disease Biology, Institute of Life Sciences, (Autonomous Institute of Department of Biotechnology, Government of India), Nalco Square, Bhubaneswar, Odisha, 751023, India
| | - Ram P Sahu
- School of Biological Sciences, National Institute of Science Education & Research, Bhubaneswar, HBNI, Jatni, Khurda, Odisha, 752050, India
| | - Chandan Goswami
- School of Biological Sciences, National Institute of Science Education & Research, Bhubaneswar, HBNI, Jatni, Khurda, Odisha, 752050, India
| | - Soma Chattopadhyay
- Infectious Disease Biology, Institute of Life Sciences, (Autonomous Institute of Department of Biotechnology, Government of India), Nalco Square, Bhubaneswar, Odisha, 751023, India.
| | - Subhasis Chattopadhyay
- School of Biological Sciences, National Institute of Science Education & Research, Bhubaneswar, HBNI, Jatni, Khurda, Odisha, 752050, India.
| |
Collapse
|
27
|
Sun Y, Nascimento Da Conceicao V, Ahamad N, Madesh M, Singh BB. Spatial localization of SOCE channels and its modulators regulate neuronal physiology and contributes to pathology. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
28
|
Guan R, Xie L, Ji L, Chao H. Phosphorescent Iridium(III) Complexes for Anticancer Applications. Eur J Inorg Chem 2020. [DOI: 10.1002/ejic.202000754] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Ruilin Guan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry Sun Yat‐Sen University 510275 Guangzhou P. R. China
| | - Lina Xie
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry Sun Yat‐Sen University 510275 Guangzhou P. R. China
| | - Liangnian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry Sun Yat‐Sen University 510275 Guangzhou P. R. China
| | - Hui Chao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry School of Chemistry Sun Yat‐Sen University 510275 Guangzhou P. R. China
- MOE Key Laboratory of Theoretical Organic Chemistry and Functional Molecule School of Chemistry and Chemical Engineering Hunan University of Science and Technology 400201 Xiangtan P. R. China
| |
Collapse
|
29
|
Li L, Venkataraman L, Chen S, Fu H. Function of WFS1 and WFS2 in the Central Nervous System: Implications for Wolfram Syndrome and Alzheimer's disease. Neurosci Biobehav Rev 2020; 118:775-783. [PMID: 32949681 DOI: 10.1016/j.neubiorev.2020.09.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/25/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022]
Abstract
L.P. Li, L. Venkataraman, S. Chen, and H.J. Fu. Function of WFS1 and WFS2 in the Central Nervous System: Implications for Wolfram Syndrome and Alzheimer's Disease. NEUROSCI BIOBEHAV REVXXX-XXX,2020.-Wolfram syndrome (WS) is a rare monogenetic spectrum disorder characterized by insulin-dependent juvenile-onset diabetes mellitus, diabetes insipidus, optic nerve atrophy, hearing loss, progressive neurodegeneration, and a wide spectrum of psychiatric manifestations. Most WS patients belong to Wolfram Syndrome type 1 (WS1) caused by mutations in the Wolfram Syndrome 1 (WFS1/Wolframin) gene, while a small fraction of patients belongs to Wolfram Syndrome type 2 (WS2) caused by pathogenic variants in the CDGSH Iron Sulfur Domain 2 (CISD2/WFS2) gene. Although currently there is no treatment for this life-threatening disease, the molecular mechanisms underlying the pathogenesis of WS have been proposed. Interestingly, Alzheimer's disease (AD), an age-dependent neurodegenerative disease, shares some common mechanisms with WS. In this review, we focus on the function of WFS1 and WFS2 in the central nervous system as well as their implications in WS and AD. We also propose three future directions for elucidating the role of WFS1 and WFS2 in WS and AD.
Collapse
Affiliation(s)
- Liangping Li
- Department of Neuroscience, Chronic Brain Injury, Discovery Themes, The Ohio State University, Columbus, OH, USA
| | - Lalitha Venkataraman
- Department of Neuroscience, Chronic Brain Injury, Discovery Themes, The Ohio State University, Columbus, OH, USA
| | - Shuo Chen
- Department of Neuroscience, Chronic Brain Injury, Discovery Themes, The Ohio State University, Columbus, OH, USA
| | - Hongjun Fu
- Department of Neuroscience, Chronic Brain Injury, Discovery Themes, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
30
|
Santoni G, Morelli MB, Marinelli O, Nabissi M, Santoni M, Amantini C. Calcium Signaling and the Regulation of Chemosensitivity in Cancer Cells: Role of the Transient Receptor Potential Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:505-517. [PMID: 31646523 DOI: 10.1007/978-3-030-12457-1_20] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer cells acquire the ability to modify the calcium signaling network by altering the expression and functions of cation channels, pumps or transporters. Calcium signaling pathways are involved in proliferation, angiogenesis, invasion, immune evasion, disruption of cell death pathways, ECM remodelling, epithelial-mesenchymal transition (EMT) and drug resistance. Among cation channels, a pivotal role is played by the Transient Receptor Potential non-selective cation-permeable receptors localized in plasma membrane, endoplasmic reticulum, mitochondria and lysosomes. Several findings indicate that the dysregulation in calcium signaling induced by TRP channels is responsible for cancer growth, metastasis and chemoresistance. Drug resistance represents a major limitation in the application of current therapeutic regimens and several efforts are spent to overcome it. Here we describe the ability of Transient Receptor Potential Channels to modify, by altering the intracellular calcium influx, the cancer cell sensitivity to chemotherapeutic drugs.
Collapse
Affiliation(s)
- Giorgio Santoni
- School of Pharmacy, Immunopathology and Molecular Medicine Laboratory, University of Camerino, Camerino, Italy
| | - Maria Beatrice Morelli
- School of Pharmacy, Immunopathology and Molecular Medicine Laboratory, University of Camerino, Camerino, Italy.,School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Oliviero Marinelli
- School of Pharmacy, Immunopathology and Molecular Medicine Laboratory, University of Camerino, Camerino, Italy.,School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Massimo Nabissi
- School of Pharmacy, Immunopathology and Molecular Medicine Laboratory, University of Camerino, Camerino, Italy
| | - Matteo Santoni
- Clinic and Oncology Unit, Macerata Hospital, Macerata, Italy
| | - Consuelo Amantini
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy.
| |
Collapse
|
31
|
|
32
|
Liu X, Zhang P, Xie C, Sham KWY, Ng SSM, Chen Y, Cheng CHK. Activation of PTEN by inhibition of TRPV4 suppresses colon cancer development. Cell Death Dis 2019; 10:460. [PMID: 31189890 PMCID: PMC6561944 DOI: 10.1038/s41419-019-1700-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 05/21/2019] [Accepted: 05/23/2019] [Indexed: 12/27/2022]
Abstract
Transient receptor potential vanilloid type 4 (TRPV4) is a Ca2+-permeable cation channel that is known to be an osmosensor and thermosensor. Currently, limited evidence shows that TRPV4 plays opposite roles in either promoting or inhibiting cancer development in different cancer types. Furthermore, the precise biological functions and the underlying mechanisms of TRPV4 in carcinogenesis are still poorly understood. In this study, we demonstrated that TRPV4 is upregulated in colon cancer and associated with poor prognosis. Contrary to the reported cell death-promoting activity of TRPV4 in certain cancer cells, TRPV4 positively regulates cell survival in human colon cancer in vitro and in vivo. Inhibition of TRPV4 affects the cell cycle progression from the G1 to S phase through modulating the protein expression of D-type cyclins. Apoptosis and autophagy induced by TRPV4 silencing attenuate cell survival and potentiate the anticancer efficacy of chemotherapeutics against colon cancer cells. In addition, PTEN is activated by inhibition of TRPV4 as indicated by the dephosphorylation and increased nuclear localization. Knockdown of PTEN significantly abrogates TRPV4 silencing induced growth inhibition and recovers the capability of clonogenicity, as well as reduced apoptosis in colon cancer cells. Thus, PTEN regulates the antigrowth effects induced by TRPV4 inhibition through both phosphatase-dependent and independent mechanisms. In conclusion, inhibition of TRPV4 suppresses colon cancer development via activation of PTEN pathway. This finding suggests that downregulation of TPRV4 expression or activity would conceivably constitute a novel approach for the treatment of human colon cancer.
Collapse
Affiliation(s)
- Xiaoyu Liu
- Longgang E.N.T. hospital & Shenzhen Key Laboratory of E.N.T., Institute of E.N.T., Shenzhen, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Peng Zhang
- Longgang E.N.T. hospital & Shenzhen Key Laboratory of E.N.T., Institute of E.N.T., Shenzhen, China
| | - Chuanming Xie
- Institute of Hepatobiliary Surgery, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing, China
| | - Kathy W Y Sham
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Simon S M Ng
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Yangchao Chen
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Christopher H K Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China.
| |
Collapse
|
33
|
Gao P, Yan Z, Zhu Z. The role of adipose TRP channels in the pathogenesis of obesity. J Cell Physiol 2019; 234:12483-12497. [PMID: 30618095 DOI: 10.1002/jcp.28106] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 12/07/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Peng Gao
- Department of Hypertension and Endocrinology Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Zhencheng Yan
- Department of Hypertension and Endocrinology Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension Chongqing China
| |
Collapse
|
34
|
Qiu K, Chen Y, Rees TW, Ji L, Chao H. Organelle-targeting metal complexes: From molecular design to bio-applications. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2017.10.022] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
35
|
Yamaguchi Y, Oh-hashi K, Matsuoka Y, Takemura H, Yamakita S, Matsuda M, Sawa T, Amaya F. Endoplasmic Reticulum Stress in the Dorsal Root Ganglion Contributes to the Development of Pain Hypersensitivity after Nerve Injury. Neuroscience 2018; 394:288-299. [DOI: 10.1016/j.neuroscience.2018.08.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 07/05/2018] [Accepted: 08/07/2018] [Indexed: 12/30/2022]
|
36
|
Prevarskaya N, Skryma R, Shuba Y. Ion Channels in Cancer: Are Cancer Hallmarks Oncochannelopathies? Physiol Rev 2018; 98:559-621. [PMID: 29412049 DOI: 10.1152/physrev.00044.2016] [Citation(s) in RCA: 277] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Genomic instability is a primary cause and fundamental feature of human cancer. However, all cancer cell genotypes generally translate into several common pathophysiological features, often referred to as cancer hallmarks. Although nowadays the catalog of cancer hallmarks is quite broad, the most common and obvious of them are 1) uncontrolled proliferation, 2) resistance to programmed cell death (apoptosis), 3) tissue invasion and metastasis, and 4) sustained angiogenesis. Among the genes affected by cancer, those encoding ion channels are present. Membrane proteins responsible for signaling within cell and among cells, for coupling of extracellular events with intracellular responses, and for maintaining intracellular ionic homeostasis ion channels contribute to various extents to pathophysiological features of each cancer hallmark. Moreover, tight association of these hallmarks with ion channel dysfunction gives a good reason to classify them as special type of channelopathies, namely oncochannelopathies. Although the relation of cancer hallmarks to ion channel dysfunction differs from classical definition of channelopathies, as disease states causally linked with inherited mutations of ion channel genes that alter channel's biophysical properties, in a broader context of the disease state, to which pathogenesis ion channels essentially contribute, such classification seems absolutely appropriate. In this review the authors provide arguments to substantiate such point of view.
Collapse
Affiliation(s)
- Natalia Prevarskaya
- INSERM U-1003, Equipe Labellisée par la Ligue Nationale contre le Cancer et LABEX, Université Lille1 , Villeneuve d'Ascq , France ; Bogomoletz Institute of Physiology and International Center of Molecular Physiology, NASU, Kyiv-24, Ukraine
| | - Roman Skryma
- INSERM U-1003, Equipe Labellisée par la Ligue Nationale contre le Cancer et LABEX, Université Lille1 , Villeneuve d'Ascq , France ; Bogomoletz Institute of Physiology and International Center of Molecular Physiology, NASU, Kyiv-24, Ukraine
| | - Yaroslav Shuba
- INSERM U-1003, Equipe Labellisée par la Ligue Nationale contre le Cancer et LABEX, Université Lille1 , Villeneuve d'Ascq , France ; Bogomoletz Institute of Physiology and International Center of Molecular Physiology, NASU, Kyiv-24, Ukraine
| |
Collapse
|
37
|
Sukumaran P, Sun Y, Zangbede FQ, da Conceicao VN, Mishra B, Singh BB. TRPC1 expression and function inhibit ER stress and cell death in salivary gland cells. FASEB Bioadv 2018; 1:40-50. [PMID: 31111119 PMCID: PMC6524637 DOI: 10.1096/fba.1021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Disturbances in endoplasmic reticulum (ER) Ca2+ homeostasis have been associated with many diseases including loss of salivary glands. Although significant progress has been accomplished which led to the increase in our understanding of the cellular responses to ER stress, the factors/ion channels that could inhibit ER stress are not yet identified. Here we show that TRPC1 (transient receptor potential canonical 1) is involved in regulating Ca2+ homeostasis and loss of TRPC1 decreased ER Ca2+ levels, inhibited the unfolded protein response (UPR), that induced loss of salivary gland cells. We provide further evidence that ER stress inducing agents (Tunicamycin and Brefeldin A) disrupts Ca2+ homeostasis by directly inhibiting TRPC1-mediated Ca2+ entry, which led to ER stress in salivary gland cells. Moreover, induction of ER stress lead to an increase in CHOP expression, which decreased TRPC1 expression and subsequently attenuated autophagy along with increased apoptosis. Importantly, TRPC1-/- mice showed increased ER stress, increased immune cell infiltration, loss of Ca2+ homeostasis, decreased saliva secretion, and decreased salivary gland survival. Finally, restoration of TRPC1 not only maintained Ca2+ homeostasis, but inhibited ER stress that induced cell survival. Overall these results suggest a significant role of TRPC1 Ca2+ channels in ER stress and homeostatic function/survival of salivary gland cells.
Collapse
Affiliation(s)
- Pramod Sukumaran
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center, San Antonio, TX 78229
| | - Yuyang Sun
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center, San Antonio, TX 78229
| | - Fredice Quenum Zangbede
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58201
| | | | - Bibhuti Mishra
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58201
| | - Brij B Singh
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center, San Antonio, TX 78229.,Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58201
| |
Collapse
|
38
|
Roest G, La Rovere RM, Bultynck G, Parys JB. IP 3 Receptor Properties and Function at Membrane Contact Sites. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 981:149-178. [PMID: 29594861 DOI: 10.1007/978-3-319-55858-5_7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) is a ubiquitously expressed Ca2+-release channel localized in the endoplasmic reticulum (ER). The intracellular Ca2+ signals originating from the activation of the IP3R regulate multiple cellular processes including the control of cell death versus cell survival via their action on apoptosis and autophagy. The exact role of the IP3Rs in these two processes does not only depend on their activity, which is modulated by the cytosolic composition (Ca2+, ATP, redox status, …) and by various types of regulatory proteins, including kinases and phosphatases as well as by a number of oncogenes and tumor suppressors, but also on their intracellular localization, especially at the ER-mitochondrial and ER-lysosomal interfaces. At these interfaces, Ca2+ microdomains are formed, in which the Ca2+ concentration is finely regulated by the different ER, mitochondrial and lysosomal Ca2+-transport systems and also depends on the functional and structural interactions existing between them. In this review, we therefore discuss the most recent insights in the role of Ca2+ signaling in general, and of the IP3R in particular, in the control of basal mitochondrial bioenergetics, apoptosis, and autophagy at the level of inter-organellar contact sites.
Collapse
Affiliation(s)
- Gemma Roest
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Leuven, Belgium
| | - Rita M La Rovere
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Leuven, Belgium
| | - Geert Bultynck
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Leuven, Belgium.
| | - Jan B Parys
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, KU Leuven, Leuven, Belgium.
| |
Collapse
|
39
|
Impaired healing of cornea incision injury in a TRPV1-deficient mouse. Cell Tissue Res 2018; 374:329-338. [PMID: 29971480 PMCID: PMC6209059 DOI: 10.1007/s00441-018-2878-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 06/21/2018] [Indexed: 12/19/2022]
Abstract
The present study attempts to elucidate the role of TRPV1 cation channel receptor on primary repair in an incision-wounded mouse cornea in vivo. Previous study revealed that blocking TRPV1 suppressed myofibroblast formation and expression of transforming growth factor β1 (TGFβ1) in cultured keratocytes or ocular fibroblasts. Male C57BL/6 (wild-type; WT) mice and male C57BL/6 Trpv1-null (KO) mice incurred a full-thickness incision injury (1.8 mm in length, limbus to limbus) in the central cornea of one eye with a surgical blade under general and topical anesthesia. The injury was not sutured. On days 0, 5, and 10, the eyes were enucleated, processed for histology, immunohistochemistry, and real-time RT-PCR gene expression analysis to evaluate the effects of the loss of TRPV1 on primary healing. Electron microscopy observation was also performed to know the effect of the loss of TRPV1 on ultrastructure of keratocytes. The results showed that the loss of Trpv1 gene delayed closure of corneal stromal incision with hindered myofibroblast transdifferentiation along with declines in the expression of collagen Ia1 and TGFβ1. Inflammatory cell infiltration was not affected by the loss of TRPV1. Ultrastructurally endoplasmic reticulum of TRPV1-null keratocytes was more extensively dilated as compared with WT keratocytes, suggesting an impairment of protein secretion by TRPV1-gene knockout. These results indicate that injury-related TRPV1 signal is involved in healing of stromal incision injury in a mouse cornea by selectively stimulating TGFβ-induced granulation tissue formation.
Collapse
|
40
|
Criollo A, Altamirano F, Pedrozo Z, Schiattarella GG, Li DL, Rivera-Mejías P, Sotomayor-Flores C, Parra V, Villalobos E, Battiprolu PK, Jiang N, May HI, Morselli E, Somlo S, de Smedt H, Gillette TG, Lavandero S, Hill JA. Polycystin-2-dependent control of cardiomyocyte autophagy. J Mol Cell Cardiol 2018. [DOI: 10.1016/j.yjmcc.2018.03.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
41
|
Ivask M, Pajusalu S, Reimann E, Kõks S. Hippocampus and Hypothalamus RNA-sequencing of WFS1-deficient Mice. Neuroscience 2018; 374:91-103. [PMID: 29406269 DOI: 10.1016/j.neuroscience.2018.01.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 11/26/2017] [Accepted: 01/18/2018] [Indexed: 01/08/2023]
Abstract
Wolfram syndrome is caused by mutations in the WFS1 gene. WFS1 protein dysfunction results in a range of neuroendocrine syndromes and is mostly characterized by juvenile-onset diabetes mellitus and optic atrophy. WFS1 has been shown to participate in membrane trafficking, protein processing and Ca2+ homeostasis in the endoplasmic reticulum. Aim of the present study was to find the transcriptomic changes influenced by WFS1 in the hypothalamus and hippocampus using RNA-sequencing. The WFS1-deficient mice were used as a model system to analyze the changes in transcriptional networks. The number of differentially expressed genes between hypothalami of WFS1-deficient (Wfs1KO) and wild-type (WT) mice was 43 and between hippocampi 311 with False Discovery Rate (FDR) <0.05. Avpr1a and Avpr1b were significantly upregulated in the hypothalamus and hippocampus of Wfs1KO mice respectively. Trpm8 was the most upregulated gene in the hippocampus of Wfs1KO mice. The functional analysis revealed significant enrichment of networks and pathways associated with protein synthesis, cell-to-cell signaling and interaction, molecular transport, metabolic disease and nervous system development and function. In conclusion, the transcriptomic profiles of WFS1-deficient hypothalamus and hippocampus do indicate the activation of degenerative molecular pathways causing the clinical occurrences typical to Wolfram syndrome.
Collapse
Affiliation(s)
- Marilin Ivask
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.
| | - Sander Pajusalu
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu, Estonia; Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Ene Reimann
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Sulev Kõks
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
42
|
Hu HJ, Song M. Disrupted Ionic Homeostasis in Ischemic Stroke and New Therapeutic Targets. J Stroke Cerebrovasc Dis 2017; 26:2706-2719. [PMID: 29054733 DOI: 10.1016/j.jstrokecerebrovasdis.2017.09.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/30/2017] [Accepted: 09/06/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Stroke is a leading cause of long-term disability. All neuroprotectants targeting excitotoxicity have failed to become stroke medications. In order to explore and identify new therapeutic targets for stroke, we here reviewed present studies of ionic transporters and channels that are involved in ischemic brain damage. METHOD We surveyed recent literature from animal experiments and clinical reports in the databases of PubMed and Elsevier ScienceDirect to analyze ionic mechanisms underlying ischemic cell damage and suggest promising ideas for stroke therapy. RESULTS Dysfunction of ionic transporters and disrupted ionic homeostasis are most early changes that underlie ischemic brain injury, thus receiving sustained attention in translational stroke research. The Na+/K+-ATPase, Na+/Ca2+ Exchanger, ionotropic glutamate receptor, acid-sensing ion channels (ASICs), sulfonylurea receptor isoform 1 (SUR1)-regulated NCCa-ATP channels, and transient receptor potential (TRP) channels are critically involved in ischemia-induced cellular degenerating processes such as cytotoxic edema, excitotoxicity, necrosis, apoptosis, and autophagic cell death. Some ionic transporters/channels also act as signalosomes to regulate cell death signaling. For acute stroke treatment, glutamate-mediated excitotoxicity must be interfered within 2 hours after stroke. The SUR1-regulated NCCa-ATP channels, Na+/K+-ATPase, ASICs, and TRP channels have a much longer therapeutic window, providing new therapeutic targets for developing feasible pharmacological treatments toward acute ischemic stroke. CONCLUSION The next generation of stroke therapy can apply a polypharmacology strategy for which drugs are designed to target multiple ion transporters/channels or their interaction with neurotoxic signaling pathways. But a successful translation of neuroprotectants relies on in-depth analyses of cell death mechanisms and suitable animal models resembling human stroke.
Collapse
Affiliation(s)
- Hui-Jie Hu
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingke Song
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
43
|
Amantini C, Farfariello V, Cardinali C, Morelli MB, Marinelli O, Nabissi M, Santoni M, Bonfili L, Cecarini V, Eleuteri AM, Santoni G. The TRPV1 ion channel regulates thymocyte differentiation by modulating autophagy and proteasome activity. Oncotarget 2017; 8:90766-90780. [PMID: 29207602 PMCID: PMC5710883 DOI: 10.18632/oncotarget.21798] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 09/20/2017] [Indexed: 12/28/2022] Open
Abstract
Autophagy and the ubiquitin-proteasome system (UPS) control thymus cell homeostasis under resting and endoplasmic reticulum (ER) stress conditions. Several evidence support a cross-talk between UPS and autophagy; abrogation of UPS responses stimulates autophagy, and vice versa the inhibition of autophagy alters the UPS functions. Herein, we found that TRPV1 activation induces ER stress, proteasome dysfunction and autophagy in thymocytes by modulating the expression of UPR-related genes. The TRPV1-mediated autophagy prevents the UPR activation by inhibiting BiP, Grp94 and ERp57 chaperone protein expression. Thymocytes from TRPV1 KO mice display both autophagy and proteasome dysfunctions, resulting in increased apoptotic cells and reduced total DP thymocyte number. In addition, positive selection of thymocytes triggered by anti-TCRβ/CD2 Ab-mediated costimulation induces apoptosis in thymocytes from TRPV1 KO as compared with WT mice. Stimulation of TRPV1 KO thymocytes with anti-TCRβ/CD2 mAbs modulates the expression of CD4 antigen on purified DP thymocytes, with reduced number of mature, single positive (SP) CD4 and increased number of immature SP CD4low and DP CD4lowCD8+ thymocytes, further supporting the intrinsic role of TRPV1 in T cell maturation. Finally, a reduction in CD8+ and CD4+ T cells is evidenced in the peripheral blood and spleen of TRPV1 KO, as compared with WT mice. Therapeutic strategy by restraining or stimulating the TRPV1 expression and functions in thymocytes might represent a new pharmacological tool in the regulation of different inflammatory T cell responses.
Collapse
Affiliation(s)
- Consuelo Amantini
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Valerio Farfariello
- University of Lille, INSERM U1003 - PHYCEL - Physiologie Cellulaire, Lille, France
| | - Claudio Cardinali
- School of Pharmacy, Experimental Medicine Section, University of Camerino, Camerino, Italy.,Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Maria Beatrice Morelli
- School of Pharmacy, Experimental Medicine Section, University of Camerino, Camerino, Italy.,Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Oliviero Marinelli
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Massimo Nabissi
- School of Pharmacy, Experimental Medicine Section, University of Camerino, Camerino, Italy
| | - Matteo Santoni
- School of Pharmacy, Experimental Medicine Section, University of Camerino, Camerino, Italy
| | - Laura Bonfili
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Valentina Cecarini
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Anna Maria Eleuteri
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Giorgio Santoni
- School of Pharmacy, Experimental Medicine Section, University of Camerino, Camerino, Italy
| |
Collapse
|
44
|
Carvacrol promotes neuroprotection in the mouse hemiparkinsonian model. Neuroscience 2017; 356:176-181. [DOI: 10.1016/j.neuroscience.2017.05.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 12/12/2022]
|
45
|
Hempel N, Trebak M. Crosstalk between calcium and reactive oxygen species signaling in cancer. Cell Calcium 2017; 63:70-96. [PMID: 28143649 DOI: 10.1016/j.ceca.2017.01.007] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/13/2017] [Accepted: 01/14/2017] [Indexed: 02/07/2023]
Abstract
The interplay between Ca2+ and reactive oxygen species (ROS) signaling pathways is well established, with reciprocal regulation occurring at a number of subcellular locations. Many Ca2+ channels at the cell surface and intracellular organelles, including the endoplasmic reticulum and mitochondria are regulated by redox modifications. In turn, Ca2+ signaling can influence the cellular generation of ROS, from sources such as NADPH oxidases and mitochondria. This relationship has been explored in great depth during the process of apoptosis, where surges of Ca2+ and ROS are important mediators of cell death. More recently, coordinated and localized Ca2+ and ROS transients appear to play a major role in a vast variety of pro-survival signaling pathways that may be crucial for both physiological and pathophysiological functions. While much work is required to firmly establish this Ca2+-ROS relationship in cancer, existing evidence from other disease models suggests this crosstalk is likely of significant importance in tumorigenesis. In this review, we describe the regulation of Ca2+ channels and transporters by oxidants and discuss the potential consequences of the ROS-Ca2+ interplay in tumor cells.
Collapse
Affiliation(s)
- Nadine Hempel
- Department of Pharmacology, Penn State College of Medicine, Hershey PA 17033, United States; Penn State Hershey Cancer Institute, Penn State College of Medicine, Hershey PA 17033, United States.
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey PA 17033, United States; Penn State Hershey Cancer Institute, Penn State College of Medicine, Hershey PA 17033, United States.
| |
Collapse
|
46
|
Molecular insights into lipid-assisted Ca 2+ regulation of the TRP channel Polycystin-2. Nat Struct Mol Biol 2017; 24:123-130. [PMID: 28092368 DOI: 10.1038/nsmb.3357] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/08/2016] [Indexed: 01/03/2023]
Abstract
Polycystin-2 (PC2), a calcium-activated cation TRP channel, is involved in diverse Ca2+ signaling pathways. Malfunctioning Ca2+ regulation in PC2 causes autosomal-dominant polycystic kidney disease. Here we report two cryo-EM structures of distinct channel states of full-length human PC2 in complex with lipids and cations. The structures reveal conformational differences in the selectivity filter and in the large exoplasmic domain (TOP domain), which displays differing N-glycosylation. The more open structure has one cation bound below the selectivity filter (single-ion mode, PC2SI), whereas multiple cations are bound along the translocation pathway in the second structure (multi-ion mode, PC2MI). Ca2+ binding at the entrance of the selectivity filter suggests Ca2+ blockage in PC2MI, and we observed density for the Ca2+-sensing C-terminal EF hand in the unblocked PC2SI state. The states show altered interactions of lipids with the pore loop and TOP domain, thus reflecting the functional diversity of PC2 at different locations, owing to different membrane compositions.
Collapse
|
47
|
Bollimuntha S, Pani B, Singh BB. Neurological and Motor Disorders: Neuronal Store-Operated Ca 2+ Signaling: An Overview and Its Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:535-556. [PMID: 28900932 PMCID: PMC5821072 DOI: 10.1007/978-3-319-57732-6_27] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Calcium (Ca2+) is a ubiquitous second messenger that performs significant physiological task such as neurosecretion, exocytosis, neuronal growth/differentiation, and the development and/or maintenance of neural circuits. An important regulatory aspect of neuronal Ca2+ homeostasis is store-operated Ca2+ entry (SOCE) which, in recent years, has gained much attention for influencing a variety of nerve cell responses. Essentially, activation of SOCE ensues following the activation of the plasma membrane (PM) store-operated Ca2+ channels (SOCC) triggered by the depletion of endoplasmic reticulum (ER) Ca2+ stores. In addition to the TRPC (transient receptor potential canonical) and the Orai family of ion channels, STIM (stromal interacting molecule) proteins have been baptized as key molecular regulators of SOCE. Functional significance of the TRPC channels in neurons has been elaborately studied; however, information on Orai and STIM components of SOCE, although seems imminent, is currently limited. Importantly, perturbations in SOCE have been implicated in a spectrum of neuropathological conditions. Hence, understanding the precise involvement of SOCC in neurodegeneration would presumably unveil avenues for plausible therapeutic interventions. We thus review the role of SOCE-regulated neuronal Ca2+ signaling in selecting neurodegenerative conditions.
Collapse
Affiliation(s)
- Sunitha Bollimuntha
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58202, USA.
| | - Biswaranjan Pani
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58202, USA
| | - Brij B Singh
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58202, USA.
| |
Collapse
|
48
|
Ma X, Gong N, Zhong L, Sun J, Liang XJ. Future of nanotherapeutics: Targeting the cellular sub-organelles. Biomaterials 2016; 97:10-21. [DOI: 10.1016/j.biomaterials.2016.04.026] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/02/2016] [Accepted: 04/20/2016] [Indexed: 11/25/2022]
|
49
|
Affiliation(s)
- Indu S Ambudkar
- Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institute of Health, Bethesda, MD 20892, USA.
| | - Shmuel Muallem
- Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institute of Health, Bethesda, MD 20892, USA
| |
Collapse
|