101
|
Wang SQ, Liu J, Qin J, Zhu Y, Tin VPC, Yam JWP, Wong MP, Xiao ZJ. CAMK2A supported tumor initiating cells of lung adenocarcinoma by upregulating SOX2 through EZH2 phosphorylation. Cell Death Dis 2020; 11:410. [PMID: 32483123 PMCID: PMC7264342 DOI: 10.1038/s41419-020-2553-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 11/10/2022]
Abstract
Tumor initiating cells (TIC) of lung cancer are mainly induced by stress-related plasticity. Calcium/Calmodulin dependent protein kinase II alpha (CAMK2A) is a key calcium signaling molecule activated by exogenous and endogenous stimuli with effects on multiple cell functions but little is known about its role on TIC. In human lung adenocarcinomas (AD), CAMK2A was aberrantly activated in a proportion of cases and was an independent risk factor predicting shorter survivals. Functionally, CAMK2A enhanced TIC phenotypes in vitro and in vivo. CAMK2A regulated SOX2 expression by reducing H3K27me3 and EZH2 occupancy at SOX2 regulatory regions, leading to its epigenetic de-repression with functional consequences. Further, CAMK2A caused kinase-dependent phosphorylation of EZH2 at T487 with suppression of EZH2 activity. Together, the data demonstrated the CAMK2A-EZH2-SOX2 axis on TIC regulation. This study provided phenotypic and mechanistic evidence for the TIC supportive role of CAMK2A, implicating a novel predictive and therapeutic target for lung cancer.
Collapse
Affiliation(s)
- Si-Qi Wang
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
| | - Jing Liu
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
| | - Jing Qin
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - Yun Zhu
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong.,Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Vicky Pui-Chi Tin
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
| | - Judy Wai Ping Yam
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
| | - Maria Pik Wong
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong
| | - Zhi-Jie Xiao
- Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong.
| |
Collapse
|
102
|
Regulation of Vascular Function and Inflammation via Cross Talk of Reactive Oxygen and Nitrogen Species from Mitochondria or NADPH Oxidase-Implications for Diabetes Progression. Int J Mol Sci 2020; 21:ijms21103405. [PMID: 32408480 PMCID: PMC7279344 DOI: 10.3390/ijms21103405] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress plays a key role for the development of cardiovascular, metabolic, and neurodegenerative disease. This concept has been proven by using the approach of genetic deletion of reactive oxygen and nitrogen species (RONS) producing, pro-oxidant enzymes as well as by the overexpression of RONS detoxifying, antioxidant enzymes leading to an amelioration of the severity of diseases. Vice versa, the development and progression of cardiovascular diseases is aggravated by overexpression of RONS producing enzymes as well as deletion of RONS detoxifying enzymes. We have previously identified cross talk mechanisms between different sources of RONS, which can amplify the oxidative stress-mediated damage. Here, the pathways and potential mechanisms leading to this cross talk are analyzed in detail and highlighted by selected examples from the current literature and own data including hypoxia, angiotensin II (AT-II)-induced hypertension, nitrate tolerance, aging, and others. The general concept of redox-based activation of RONS sources via “kindling radicals” and enzyme-specific “redox switches” as well as the interaction with redox-sensitive inflammatory pathways are discussed. Here, we present evidence for the existence of such cross talk mechanisms in the setting of diabetes and critically assess their contribution to the severity of diabetic complications.
Collapse
|
103
|
The mystery of mitochondria-ER contact sites in physiology and pathology: A cancer perspective. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165834. [PMID: 32437958 DOI: 10.1016/j.bbadis.2020.165834] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/29/2020] [Accepted: 05/01/2020] [Indexed: 12/13/2022]
Abstract
Mitochondria-associated membranes (MAM), physical platforms that enable communication between mitochondria and the endoplasmic reticulum (ER), are enriched with many proteins and enzymes involved in several crucial cellular processes, such as calcium (Ca2+) homeostasis, lipid synthesis and trafficking, autophagy and reactive oxygen species (ROS) production. Accumulating studies indicate that tumor suppressors and oncogenes are present at these intimate contacts between mitochondria and the ER, where they influence Ca2+ flux between mitochondria and the ER or affect lipid homeostasis at MAM, consequently impacting cell metabolism and cell fate. Understanding these fundamental roles of mitochondria-ER contact sites as important domains for tumor suppressors and oncogenes can support the search for new and more precise anticancer therapies. In the present review, we summarize the current understanding of basic MAM biology, composition and function and discuss the possible role of MAM-resident oncogenes and tumor suppressors.
Collapse
|
104
|
Sies H, Jones DP. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat Rev Mol Cell Biol 2020; 21:363-383. [PMID: 32231263 DOI: 10.1038/s41580-020-0230-3] [Citation(s) in RCA: 2431] [Impact Index Per Article: 486.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2020] [Indexed: 02/07/2023]
Abstract
'Reactive oxygen species' (ROS) is an umbrella term for an array of derivatives of molecular oxygen that occur as a normal attribute of aerobic life. Elevated formation of the different ROS leads to molecular damage, denoted as 'oxidative distress'. Here we focus on ROS at physiological levels and their central role in redox signalling via different post-translational modifications, denoted as 'oxidative eustress'. Two species, hydrogen peroxide (H2O2) and the superoxide anion radical (O2·-), are key redox signalling agents generated under the control of growth factors and cytokines by more than 40 enzymes, prominently including NADPH oxidases and the mitochondrial electron transport chain. At the low physiological levels in the nanomolar range, H2O2 is the major agent signalling through specific protein targets, which engage in metabolic regulation and stress responses to support cellular adaptation to a changing environment and stress. In addition, several other reactive species are involved in redox signalling, for instance nitric oxide, hydrogen sulfide and oxidized lipids. Recent methodological advances permit the assessment of molecular interactions of specific ROS molecules with specific targets in redox signalling pathways. Accordingly, major advances have occurred in understanding the role of these oxidants in physiology and disease, including the nervous, cardiovascular and immune systems, skeletal muscle and metabolic regulation as well as ageing and cancer. In the past, unspecific elimination of ROS by use of low molecular mass antioxidant compounds was not successful in counteracting disease initiation and progression in clinical trials. However, controlling specific ROS-mediated signalling pathways by selective targeting offers a perspective for a future of more refined redox medicine. This includes enzymatic defence systems such as those controlled by the stress-response transcription factors NRF2 and nuclear factor-κB, the role of trace elements such as selenium, the use of redox drugs and the modulation of environmental factors collectively known as the exposome (for example, nutrition, lifestyle and irradiation).
Collapse
Affiliation(s)
- Helmut Sies
- Institute for Biochemistry and Molecular Biology I, Heinrich Heine University Düsseldorf, Düsseldorf, Germany. .,Leibniz Research Institute for Environmental Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| | - Dean P Jones
- Department of Medicine, Emory University, Atlanta, GA, USA.
| |
Collapse
|
105
|
Wang DP, Wang ZJ, Zhao R, Lin CX, Sun QY, Yan CP, Zhou X, Cao JM. Silica nanomaterials induce organ injuries by Ca 2+-ROS-initiated disruption of the endothelial barrier and triggering intravascular coagulation. Part Fibre Toxicol 2020; 17:12. [PMID: 32293491 PMCID: PMC7087393 DOI: 10.1186/s12989-020-00340-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 02/06/2020] [Indexed: 11/23/2022] Open
Abstract
Background The growing use of silica nanoparticles (SiNPs) in many fields raises human toxicity concerns. We studied the toxicity of SiNP-20 (particle diameter 20 nm) and SiNP-100 (100 nm) and the underlying mechanisms with a focus on the endothelium both in vitro and in vivo. Methods The study was conducted in cultured human umbilical vein endothelial cells (HUVECs) and adult female Balb/c mice using several techniques. Results In vitro, both SiNP-20 and SiNP-100 decreased the viability and damaged the plasma membrane of cultured HUVECs. The nanoparticles also inhibited HUVECs migration and tube formation in a concentration-dependent manner. Both SiNPs induced significant calcium mobilization and generation of reactive oxygen species (ROS), increased the phosphorylation of vascular endothelial (VE)-cadherin at the site of tyrosine 731 residue (pY731-VEC), decreased the expression of VE-cadherin expression, disrupted the junctional VE-cadherin continuity and induced F-actin re-assembly in HUVECs. The injuries were reversed by blocking Ca2+ release activated Ca2+ (CRAC) channels with YM58483 or by eliminating ROS with N-acetyl cysteine (NAC). In vivo, both SiNP-20 and SiNP-100 (i.v.) induced multiple organ injuries of Balb/c mice in a dose (range 7–35 mg/kg), particle size, and exposure time (4–72 h)-dependent manner. Heart injuries included coronary endothelial damage, erythrocyte adhesion to coronary intima and coronary coagulation. Abdominal aorta injury exhibited intimal neoplasm formation. Lung injuries were smaller pulmonary vein coagulation, bronchiolar epithelial edema and lumen oozing and narrowing. Liver injuries included multifocal necrosis and smaller hepatic vein congestion and coagulation. Kidney injuries involved glomerular congestion and swelling. Macrophage infiltration occurred in all of the observed organ tissues after SiNPs exposure. SiNPs also decreased VE-cadherin expression and altered VE-cadherin spatial distribution in multiple organ tissues in vivo. The largest SiNP (SiNP-100) and longest exposure time exerted the greatest toxicity both in vitro and in vivo. Conclusions SiNPs, administrated in vivo, induced multiple organ injuries, including endothelial damage, intravascular coagulation, and secondary inflammation. The injuries are likely caused by upstream Ca2+-ROS signaling and downstream VE-cadherin phosphorylation and destruction and F-actin remodeling. These changes led to endothelial barrier disruption and triggering of the contact coagulation pathway.
Collapse
Affiliation(s)
- De-Ping Wang
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Zhao-Jun Wang
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Rong Zhao
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Cai-Xia Lin
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Qian-Yu Sun
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Cai-Ping Yan
- Center of Translational Medicine, Shanxi Medical University, Taiyuan, China
| | - Xin Zhou
- Department of Medical Imaging, Shanxi Medical University, Taiyuan, China.
| | - Ji-Min Cao
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
106
|
Ivanova DG, Yaneva ZL. Antioxidant Properties and Redox-Modulating Activity of Chitosan and Its Derivatives: Biomaterials with Application in Cancer Therapy. Biores Open Access 2020; 9:64-72. [PMID: 32219012 PMCID: PMC7097683 DOI: 10.1089/biores.2019.0028] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Many studies have shown that mitochondrial metabolism has a fundamental role in induction of carcinogenesis due to the influence of increased levels of reactive oxygen species (ROS) generation in all steps of oncogene transformation and cancer progression. It is widely accepted that the anticancer effect of conventional anticancer drugs is due to induction of oxidative stress and elevated intracellular levels of ROS, which alter the redox homeostasis of cancer cells. On the other hand, the harmful side effects of conventional anticancer chemotherapeutics are also due to increased production of ROS and disruption of redox homeostasis of normal cells and tissues. Therefore, there is a growing interest toward the development of natural antioxidant compounds from various sources, which could impact the redox state of cancer and normal cells by different pathways and could prevent damage from oxidant-mediated reactions. It is known that chitosan exhibits versatile biological properties, including biodegradability, biocompatibility, and a less toxic nature. Because of its antioxidant, antibacterial, anticancer, anti-inflammatory, and immunostimulatory activities, the biopolymer has been used in a wide variety of pharmaceutical, biomedical, food industry, health, and agricultural applications and has been classified as a new physiologically bioactive material.
Collapse
Affiliation(s)
- Donika G. Ivanova
- Department of Pharmacology, Animal Physiology and Physiology Chemistry, Trakia University, Stara Zagora, Bulgaria
| | - Zvezdelina L. Yaneva
- Department of Pharmacology, Animal Physiology and Physiology Chemistry, Trakia University, Stara Zagora, Bulgaria
| |
Collapse
|
107
|
Mohanty S, Barik P, Debata N, Nagarajan P, Devadas S. iCa 2+ Flux, ROS and IL-10 Determines Cytotoxic, and Suppressor T Cell Functions in Chronic Human Viral Infections. Front Immunol 2020; 11:83. [PMID: 32210950 PMCID: PMC7068714 DOI: 10.3389/fimmu.2020.00083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 01/13/2020] [Indexed: 12/13/2022] Open
Abstract
Exhaustion of CD8+ T cells and increased IL-10 production is well-known in chronic viral infections but mechanisms leading to loss of their cytotoxic capabilities and consequent exhaustion remain unclear. Exhausted CD8+T cells also called T suppressors are highly immune suppressive with altered T cell receptor signaling characteristics that mark it exclusively from their cytotoxic counterparts. Our study found that iCa2+ flux is reduced following T cell receptor activation in T suppressor cells when compared to their effector counterpart. Importantly chronic activation of murine cytotoxic CD8+ T cells lead to reduced iCa2+ influx, decreased IFN-γ and enhanced IL-10 production and this profile is mimicked in Tc1 cells upon reduction of iCa2+ flux by extracellular calcium channel inhibitors. Further reduced iCa2+ flux induced ROS which lead to IFN-γ reduction and increased IL-10 producing T suppressors through the STAT3—STAT5 axis. The above findings were substantiated by our human data where reduced iCa2+ flux in chronic Hepatitis infections displayed CD8+ T cells with low IFN-γ and increased IL-10 production. Importantly treatment with an antioxidant led to increased IFN-γ and reduced IL-10 production in human chronic Hep-B/C samples suggesting overall a proximal regulatory role for iCa2+ influx, ROS, and IL-10 in determining the effector/ suppressive axis of CD8+ T cells.
Collapse
Affiliation(s)
- Subhasmita Mohanty
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, India
| | - Prakash Barik
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, India
| | - Nagen Debata
- Department of Pathology, Institute of Medical Sciences and SUM Hospital, Bhubaneswar, India
| | - Perumal Nagarajan
- Experimental Animal Facility, National Institute of Immunology, New Delhi, India
| | - Satish Devadas
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, India
| |
Collapse
|
108
|
Xu Z, Han S, Gu Z, Wu J. Advances and Impact of Antioxidant Hydrogel in Chronic Wound Healing. Adv Healthc Mater 2020; 9:e1901502. [PMID: 31977162 DOI: 10.1002/adhm.201901502] [Citation(s) in RCA: 343] [Impact Index Per Article: 68.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/15/2019] [Indexed: 01/20/2023]
Abstract
The accelerating and thorough treatment of chronic wounds still represents a major unmet medical need owing to the complex symptoms resulting from metabolic disorder of the wound microenvironment. Although numerous strategies and bioactive hydrogels are developed, an effective and widely used method of chronic wound treatment remains a bottleneck. With the aim to accelerate chronic wound healing, many hydrogel dressings with antioxidant functions have emerged and are proven to accelerate wound healing, especially for chronic wound repair. The new strategy in chronic wound treatment brought by antioxidant hydrogels is of great significance to human health. Here, the application of antioxidant hydrogels in the repair of chronic wounds is discussed systematically, aiming to provide an important theoretical reference for the further breakthrough of chronic wound healing.
Collapse
Affiliation(s)
- Zejun Xu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong ProvinceSchool of Biomedical EngineeringSun Yat‐sen University Guangzhou 510006 P. R. China
| | - Shuyan Han
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong ProvinceSchool of Biomedical EngineeringSun Yat‐sen University Guangzhou 510006 P. R. China
| | - Zhipeng Gu
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials EngineeringSichuan University Chengdu 610065 P. R. China
- Research Institute of Sun Yat‐sen University in Shenzhen Shenzhen 518057 P. R. China
| | - Jun Wu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong ProvinceSchool of Biomedical EngineeringSun Yat‐sen University Guangzhou 510006 P. R. China
- Research Institute of Sun Yat‐sen University in Shenzhen Shenzhen 518057 P. R. China
| |
Collapse
|
109
|
Characterization of cytotoxic effects of aristolochic acids on the vascular endothelium. Toxicol In Vitro 2020; 65:104811. [PMID: 32119997 DOI: 10.1016/j.tiv.2020.104811] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/09/2020] [Accepted: 02/23/2020] [Indexed: 02/08/2023]
Abstract
Aristolochic acid nephropathy (AAN) is characterized by interstitial fibrosis, proximal tubular atrophy, and hypoxia. A correlation between a reduced peritubular capillary density and the severity of fibrosis has been demonstrated. As calcium, redox and energetic homeostasis are crucial in maintaining endothelial cell function and survival, we aimed to investigate AA-induced disturbances involved in endothelial cell injury. Our results showed a cytotoxic effect of AA on EAhy926 endothelial cells. Exposure of aortic rings to AA impaired vascular relaxation to Acetylcholine (ACh). Increased levels of intracellular reactive oxygen species (ROS) were observed in cells exposed to AA. Pre-treatment with antioxidant N-acetyl cysteine inhibited AA-induced cell death. Superoxide dismutase resulted in restoring ACh-induced relaxation. An increase in intracellular calcium level ([Ca2+]i) was observed on endothelial cells. Calcium chelators BAPTA-AM or APB, a specific inhibitor of IP3R, improved cell viability. Moreover, AA exposure led to reduced AMP-activated protein kinase (AMPK) expression. AICAR, an activator of AMPK, improved the viability of AA-intoxicated cells and inhibited the rise of cytosolic [Ca2+]i levels. This study provides evidence that AA exposure increases ROS generation, disrupts calcium homeostasis and decreases AMPK activity. It also suggests that significant damage observed in endothelial cells may enhance microcirculation defects, worsening hypoxia and tubulointerstitial lesions.
Collapse
|
110
|
Mitochondrial Calcium Regulation of Redox Signaling in Cancer. Cells 2020; 9:cells9020432. [PMID: 32059571 PMCID: PMC7072435 DOI: 10.3390/cells9020432] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/05/2020] [Accepted: 02/10/2020] [Indexed: 02/07/2023] Open
Abstract
Calcium (Ca2+) uptake into the mitochondria shapes cellular Ca2+ signals and acts as a key effector for ATP generation. In addition, mitochondria-derived reactive oxygen species (mROS), produced as a consequence of ATP synthesis at the electron transport chain (ETC), modulate cellular signaling pathways that contribute to many cellular processes. Cancer cells modulate mitochondrial Ca2+ ([Ca2+]m) homeostasis by altering the expression and function of mitochondrial Ca2+ channels and transporters required for the uptake and extrusion of mitochondrial Ca2+. Regulated elevations in [Ca2+]m are required for the activity of several mitochondrial enzymes, and this in turn regulates metabolic flux, mitochondrial ETC function and mROS generation. Alterations in both [Ca2+]m and mROS are hallmarks of many tumors, and elevated mROS is a known driver of pro-tumorigenic redox signaling, resulting in the activation of pathways implicated in cellular proliferation, metabolic alterations and stress-adaptations. In this review, we highlight recent studies that demonstrate the interplay between [Ca2+]m and mROS signaling in cancer.
Collapse
|
111
|
Oono K, Ohtake K, Watanabe C, Shiba S, Sekiya T, Kasono K. Contribution of Pyk2 pathway and reactive oxygen species (ROS) to the anti-cancer effects of eicosapentaenoic acid (EPA) in PC3 prostate cancer cells. Lipids Health Dis 2020; 19:15. [PMID: 32005121 PMCID: PMC6993438 DOI: 10.1186/s12944-019-1122-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 09/30/2019] [Indexed: 01/04/2023] Open
Abstract
Background n-3 polyunsaturated fatty acids (n-3 PUFAs), including eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are thought to exert protective effects in cardiovascular diseases. In addition, n-3 PUFAs have demonstrated anti-cancer effects in vitro and in vivo. Objective We investigated the anti-cancer effects and mechanism of action of EPA on PC3 prostate cancer cells in vitro. Methods PC3 cells were treated with various concentrations of EPA, and cell survival and the abilities of migration and invasion were evaluated. The time course of the growth inhibitory effect of EPA on PC3 cells was also assessed. The mechanism underlying the anti-cancer effects of EPA was investigated by human phosphokinase and human apoptosis antibody arrays, and confirmed by western blot analysis. We also examined the contribution of reactive oxygen species (ROS) to the effects of EPA using the ROS inhibitor N-acetyl cysteine. Results EPA decreased the survival of PC3 cells in a dose-dependent manner within 3 h of application, with an effective concentration of 500 μmol/L. EPA inhibited proline-rich tyrosine kinase (Pyk)2 and extracellular signal-regulated kinase 1/2 phosphorylation as determined by western blotting and the antibody arrays. The growth of PC3 cells was inhibited by EPA, which was dependent on ROS induction, while EPA inhibited Pyk2 phosphorylation independent of ROS production. Conclusions Inhibition of Pyk2 phosphorylation and ROS production contribute to the anticancer effects of EPA on PC3 cells.
Collapse
Affiliation(s)
- Keiichi Oono
- Laboratory of Physiology, Faculty of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Kazuo Ohtake
- Laboratory of Physiology, Faculty of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Chie Watanabe
- Laboratory of Clinical Pathology, Faculty of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Sachiko Shiba
- Laboratory of Physiology, Faculty of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Takashi Sekiya
- Laboratory of Clinical Pathology, Faculty of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Keizo Kasono
- Laboratory of Physiology, Faculty of Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan.
| |
Collapse
|
112
|
Pathak T, Gueguinou M, Walter V, Delierneux C, Johnson MT, Zhang X, Xin P, Yoast RE, Emrich SM, Yochum GS, Sekler I, Koltun WA, Gill DL, Hempel N, Trebak M. Dichotomous role of the human mitochondrial Na +/Ca2 +/Li + exchanger NCLX in colorectal cancer growth and metastasis. eLife 2020; 9:59686. [PMID: 32914752 PMCID: PMC7529464 DOI: 10.7554/elife.59686] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/07/2020] [Indexed: 12/28/2022] Open
Abstract
Despite the established role of mitochondria in cancer, the mechanisms by which mitochondrial Ca2+ (mtCa2+) regulates tumorigenesis remain incompletely understood. The crucial role of mtCa2+ in tumorigenesis is highlighted by altered expression of proteins mediating mtCa2+ uptake and extrusion in cancer. Here, we demonstrate decreased expression of the mitochondrial Na+/Ca2+/Li+ exchanger NCLX (SLC8B1) in human colorectal tumors and its association with advanced-stage disease in patients. Downregulation of NCLX causes mtCa2+ overload, mitochondrial depolarization, decreased expression of cell-cycle genes and reduced tumor size in xenograft and spontaneous colorectal cancer mouse models. Concomitantly, NCLX downregulation drives metastatic spread, chemoresistance, and expression of epithelial-to-mesenchymal, hypoxia, and stem cell pathways. Mechanistically, mtCa2+ overload leads to increased mitochondrial reactive oxygen species, which activate HIF1α signaling supporting metastasis of NCLX-null tumor cells. Thus, loss of NCLX is a novel driver of metastasis, indicating that regulation of mtCa2+ is a novel therapeutic approach in metastatic colorectal cancer.
Collapse
Affiliation(s)
- Trayambak Pathak
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of MedicineHersheyUnited States
| | - Maxime Gueguinou
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of MedicineHersheyUnited States
| | - Vonn Walter
- Department of Public Health Sciences, The Pennsylvania State University College of MedicineHersheyUnited States,Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of MedicineHersheyUnited States,Penn State Cancer Institute. The Pennsylvania State University College of MedicineHersheyUnited States
| | - Celine Delierneux
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of MedicineHersheyUnited States
| | - Martin T Johnson
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of MedicineHersheyUnited States
| | - Xuexin Zhang
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of MedicineHersheyUnited States
| | - Ping Xin
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of MedicineHersheyUnited States
| | - Ryan E Yoast
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of MedicineHersheyUnited States
| | - Scott M Emrich
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of MedicineHersheyUnited States
| | - Gregory S Yochum
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of MedicineHersheyUnited States,Department of Surgery, Division of Colon and Rectal Surgery, The Pennsylvania State University College of MedicineHersheyUnited States
| | - Israel Sekler
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the NegevBeer ShevaIsrael
| | - Walter A Koltun
- Department of Surgery, Division of Colon and Rectal Surgery, The Pennsylvania State University College of MedicineHersheyUnited States
| | - Donald L Gill
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of MedicineHersheyUnited States
| | - Nadine Hempel
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of MedicineHersheyUnited States,Penn State Cancer Institute. The Pennsylvania State University College of MedicineHersheyUnited States,Department of Pharmacology, The Pennsylvania State University College of MedicineHersheyUnited States
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of MedicineHersheyUnited States
| |
Collapse
|
113
|
Sakaguchi R, Mori Y. Transient receptor potential (TRP) channels: Biosensors for redox environmental stimuli and cellular status. Free Radic Biol Med 2020; 146:36-44. [PMID: 31682917 DOI: 10.1016/j.freeradbiomed.2019.10.415] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 10/26/2019] [Accepted: 10/29/2019] [Indexed: 12/26/2022]
Abstract
Transient receptor potential (TRP) channels are a family of cation channels that depolarizes the membrane potential and regulates intracellular concentrations of cations such as Ca2+. TRP channels are also known to function as "biosensors" to detect changes of the surrounding environment and cellular status. Lines of evidence have unveiled that numerous proteins are subject to redox modification and subsequent signaling. For example, TRPM2, TRPC5, TRPV1, and TRPA1 are known as redox sensors activated by hydrogen peroxide (H2O2), nitric oxide (NO), and electrophiles. Thus, these channels facilitate the influx of cations which in turn triggers the appropriate cellular responses against environmental redox stimuli and cellular redox status. In this review, we focus on the recent findings regarding the functions of TRP channels in relation to other ion channels, and other proteins which also go through redox modification of cysteine (Cys) residues. We aim to understand the structural and molecular basis of the redox-sensing mechanisms of TRP channels in exerting various functions under physiological conditions as well as pathological conditions such as cancer malignancy. Their future potential as drug targets will also be discussed.
Collapse
Affiliation(s)
- Reiko Sakaguchi
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan; The World Premier International Research Initiative-Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, 615-8510, Japan
| | - Yasuo Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, 615-8510, Japan; The World Premier International Research Initiative-Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, 615-8510, Japan.
| |
Collapse
|
114
|
Tsubata T. Involvement of Reactive Oxygen Species (ROS) in BCR Signaling as a Second Messenger. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1254:37-46. [PMID: 32323267 DOI: 10.1007/978-981-15-3532-1_3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) are not only toxic substances inducing oxidative stress but also play a role in receptor signaling as a second messenger, which augments signaling through various receptors by oxidizing ROS-sensitive signaling molecules. Among ROS, H2O2 is suggested to be an important second messenger because of its relative stability. H2O2 is generated by superoxide dismutase (SOD)-mediated conversion of superoxide produced by membrane-localized NADPH oxidases (NOXes). Superoxide and H2O2 are also produced as a by-product of mitochondrial respiratory chain and various other metabolic reactions. BCR ligation induces ROS production in two phases. ROS production starts immediately after BCR ligation and ceases in 1 h, then re-starts 2 h after BCR ligation and lasts 4-6 h. ROS production in the early phase is mediated by NOX2, a NOX isoform, but does not regulate BCR signaling. In contrast, ROS production at the late phase augments BCR signaling. Although the involvement of mitochondrial respiration was previously suggested in prolonged BCR ligation-induced ROS production, we recently demonstrated that NOX3, another NOX isoform, plays a central role in ROS production at the late phase. NOXes are shown to be a component of ROS-generating signaling endosome called redoxosome together with endocytosed receptors and receptor-associated signaling molecules. In redoxosome, ROS generated by NOXes augment signaling through the endocytosed receptor. The role of NOXes and redoxosome in BCR signaling needs to be further elucidated.
Collapse
Affiliation(s)
- Takeshi Tsubata
- Department of Immunology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan.
| |
Collapse
|
115
|
Wu F, Xiong H, Sha S. Noise-induced loss of sensory hair cells is mediated by ROS/AMPKα pathway. Redox Biol 2019; 29:101406. [PMID: 31926629 PMCID: PMC6933152 DOI: 10.1016/j.redox.2019.101406] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/25/2019] [Accepted: 12/09/2019] [Indexed: 12/25/2022] Open
Abstract
The formation of reactive oxygen species (ROS) is a well-documented process in noise-induced hearing loss (NIHL). We have also previously shown that activation of 5' adenosine monophosphate (AMP)-activated protein kinase (AMPKα) at its catalytic residue T172 is one of the key reactions triggering noise-induced outer hair cell (OHC) death. In this study, we are addressing the link between ROS formation and activation of AMPKα in OHCs after noise exposure. In-vivo treatment of CBA/J mice with the antioxidant N-acetyl cysteine (NAC) reduced noise-induced ROS formation (as assessed by the relative levels of 4-hydroxynonenal and 3-nitrotyrosine) and activation of AMPKα in OHCs. Forskolin, an activator of adenylyl cyclase (AC) and an antioxidant, significantly increased cyclic adenosine monophosphate (cAMP) and decreased ROS formation and noise-induced activation of AMPKα. Consequently, treatment with forskolin attenuated noise-induced losses of OHCs and NIHL. In HEI-OC1 cells, H2O2-induced activation of AMPKα and cell death were inhibited by the application of forskolin. The sum of our data indicates that noise activates AMPKα in OHCs through formation of ROS and that noise-exposure-induced OHC death is mediated by a ROS/AMPKα-dependent pathway. Forskolin may serve as a potential compound for prevention of NIHL.
Collapse
Affiliation(s)
- Fan Wu
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA; Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hao Xiong
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA; Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Suhua Sha
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
116
|
Dou JP, Wu Q, Fu CH, Zhang DY, Yu J, Meng XW, Liang P. Amplified intracellular Ca 2+ for synergistic anti-tumor therapy of microwave ablation and chemotherapy. J Nanobiotechnology 2019; 17:118. [PMID: 31791353 PMCID: PMC6889637 DOI: 10.1186/s12951-019-0549-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 11/18/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Developing new strategies to reduce the output power of microwave (MW) ablation while keeping anti-tumor effect are highly desirable for the simultaneous achievement of effective tumor killing and avoidance of complications. We find that mild MW irradiation can significantly increase intracellular Ca2+ concentration in the presence of doxorubicin hydrochloride (DOX) and thus induce massive tumor cell apoptosis. Herein, we designed a synergistic nanoplatform that not only amplifies the intracellular Ca2+ concentration and induce cell death under mild MW irradiation but also avoids the side effect of thermal ablation and chemotherapy. RESULTS The as-made NaCl-DOX@PLGA nanoplatform selectively elevates the temperature of tumor tissue distributed with nanoparticles under low-output MW, which further prompts the release of DOX from the PLGA nanoparticles and tumor cellular uptake of DOX. More importantly, its synergistic effect not only combines thermal ablation and chemotherapy, but also obviously increases the intracellular Ca2+ concentration. Changes of Ca2+ broke the homeostasis of tumor cells, decreased the mitochondrial inner membrane potential and finally induced the cascade of apoptosis under nonlethal temperature. As such, the NaCl-DOX@PLGA efficiently suppressed the tumor cell progression in vivo and in vitro under mild MW irradiation for the triple synergic effect. CONCLUSIONS This work provides a biocompatible and biodegradable nanoplatform with triple functions to realize the effective tumor killing in unlethal temperature. Those findings provide reliable solution to solve the bottleneck problem bothering clinics about the balance of thermal efficiency and normal tissue protection.
Collapse
Affiliation(s)
- Jian-Ping Dou
- Department of Interventional Ultrasound, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Qiong Wu
- Laboratory of Controllable Preparation and Application of Nanomaterials, CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, People's Republic of China
| | - Chang-Hui Fu
- Laboratory of Controllable Preparation and Application of Nanomaterials, CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, People's Republic of China
| | - Dong-Yun Zhang
- Department of Interventional Ultrasound, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Jie Yu
- Department of Interventional Ultrasound, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Xian-Wei Meng
- Laboratory of Controllable Preparation and Application of Nanomaterials, CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, People's Republic of China.
| | - Ping Liang
- Department of Interventional Ultrasound, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
117
|
Fibroblasts to Keratinocytes Redox Signaling: The Possible Role of ROS in Psoriatic Plaque Formation. Antioxidants (Basel) 2019; 8:antiox8110566. [PMID: 31752190 PMCID: PMC6912201 DOI: 10.3390/antiox8110566] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/08/2019] [Accepted: 11/12/2019] [Indexed: 12/27/2022] Open
Abstract
Although the role of reactive oxygen species-mediated (ROS-mediated) signalling in physiologic and pathologic skin conditions has been proven, no data exist on the skin cells ROS-mediated communication. Primary fibroblasts were obtained from lesional and non-lesional skin of psoriatic patients. ROS, superoxide anion, calcium and nitric oxide levels and lipoperoxidation markers and total antioxidant content were measured in fibroblasts. NADPH oxidase activity and NOX1, 2 and 4 expressions were assayed and NOX4 silencing was performed. Fibroblasts and healthy keratinocytes co-culture was performed. MAPK pathways activation was studied in fibroblasts and in co-cultured healthy keratinocytes. Increased intracellular calcium, •NO and ROS levels as well as an enhanced NADPH oxidase 4 (NOX4)-mediated extracellular ROS release was shown in lesional psoriatic vs. control fibroblasts. Upon co-culture with lesional fibroblasts, keratinocytes showed p38 and ERK MAPKs pathways activation, ROS, Ca2+ and •NO increase and cell cycle acceleration. Notably, NOX4 knockdown significantly reduced the observed effects of lesional fibroblasts on keratinocyte cell cycle progression. Co-culture with non-lesional psoriatic and control fibroblasts induced slight cell cycle acceleration, but notable intracellular ROS accumulation and ERK MAPK activation in keratinocytes. Collectively, our data demonstrate that NOX4 expressed in dermal fibroblasts is essential for the redox paracrine regulation of epidermal keratinocytes proliferation.
Collapse
|
118
|
Zhang X, Lee MD, Wilson C, McCarron JG. Hydrogen peroxide depolarizes mitochondria and inhibits IP 3-evoked Ca 2+ release in the endothelium of intact arteries. Cell Calcium 2019; 84:102108. [PMID: 31715384 PMCID: PMC6891240 DOI: 10.1016/j.ceca.2019.102108] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/30/2019] [Accepted: 10/30/2019] [Indexed: 12/30/2022]
Abstract
H2O2 is produced by several cell processes including mitochondria and may act as an intracellular messenger and cell-cell signalling molecule. Spontaneous local Ca2+ signals and IP3-evoked Ca2+ increases were inhibited by H2O2. H2O2 suppression of IP3-evoked Ca2+ signalling may be mediated by mitochondria via a decrease in the mitochondrial membrane potential. H2O2-induced mitochondrial depolarization and inhibition of IP3-evoked Ca2+ release, may protect mitochondria from Ca2+ overload during IP3-linked Ca2+ signals.
Hydrogen peroxide (H2O2) is a mitochondrial-derived reactive oxygen species (ROS) that regulates vascular signalling transduction, vasocontraction and vasodilation. Although the physiological role of ROS in endothelial cells is acknowledged, the mechanisms underlying H2O2 regulation of signalling in native, fully-differentiated endothelial cells is unresolved. In the present study, the effects of H2O2 on Ca2+ signalling were investigated in the endothelium of intact rat mesenteric arteries. Spontaneous local Ca2+ signals and acetylcholine evoked Ca2+ increases were inhibited by H2O2. H2O2 inhibition of acetylcholine-evoked Ca2+ signals was reversed by catalase. H2O2 exerts its inhibition on the IP3 receptor as Ca2+ release evoked by photolysis of caged IP3 was supressed by H2O2. H2O2 suppression of IP3-evoked Ca2+ signalling may be mediated by mitochondria. H2O2 depolarized mitochondria membrane potential. Acetylcholine-evoked Ca2+ release was inhibited by depolarisation of the mitochondrial membrane potential by the uncoupler carbonyl cyanide 3-chlorophenylhydrazone (CCCP) or complex 1 inhibitor, rotenone. We propose that the suppression of IP3-evoked Ca2+ release by H2O2 arises from the decrease in mitochondrial membrane potential. These results suggest that mitochondria may protect themselves against Ca2+ overload during IP3-linked Ca2+ signals by a H2O2 mediated negative feedback depolarization of the organelle and inhibition of IP3-evoked Ca2+ release.
Collapse
Affiliation(s)
- Xun Zhang
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Matthew D Lee
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Calum Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - John G McCarron
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| |
Collapse
|
119
|
Qian B, Wei L, Yang Z, He Q, Chen H, Wang A, Yang D, Li Q, Li J, Zheng S, Fu W. Hic-5 in pancreatic stellate cells affects proliferation, apoptosis, migration, invasion of pancreatic cancer cells and postoperative survival time of pancreatic cancer. Biomed Pharmacother 2019; 121:109355. [PMID: 31683179 DOI: 10.1016/j.biopha.2019.109355] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 08/07/2019] [Accepted: 08/14/2019] [Indexed: 12/26/2022] Open
Abstract
Pancreatic cancer is one of the most severe types of tumors, with a 5-year survival rate of less than 7%. The prognosis and treatment of pancreatic cancer are largely limited by the extent of tumor invasion and the presence of lymph node and distant metastases. Therefore, exploring the biological behavior of pancreatic cancer cells (PCCs) is extremely important for the understanding, diagnosis, and treatment of pancreatic cancer. Current studies have shown that pancreatic stellate cells (PSCs) regulate the biological behavior of PCCs, such as their proliferation, apoptosis, invasion, and migration, by remodeling the extracellular matrix. Though Hic-5 is an important gene in PSCs, no study has investigated the regulation of PCCs by Hic-5. Here, we demonstrate that Hic-5 expression is upregulated in pancreatic cancer and that siRNA transfection can effectively inhibit Hic-5 expression. Compared to the control group, Hic-5 inhibition significantly reduced proliferation, increased apoptosis, and reduced invasion and migration of PCCs. Moreover, the inhibition of Hic-5 expression simultaneously reduced matrix metalloproteinase-9 (MMP-9) expression. Statistical analysis revealed that Hic-5 expression was higher among the pancreatic cancer group than among the normal group and was negatively correlated with postoperative survival time among patients with pancreatic cancer. These results have important clinical significance for further exploring the molecular mechanism involved in Hic-5-mediated invasion and metastasis of pancreatic cancer and ameliorating the prognosis of patients with pancreatic cancer.
Collapse
Affiliation(s)
- Baolin Qian
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Liping Wei
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Zhongqiu Yang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qinyuan He
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Hao Chen
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Ankang Wang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Dayin Yang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qiu Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jing Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Silin Zheng
- Department of Nursing, The Affiliated Hospital of Southwest Medical University, Sichuan, China.
| | - Wenguang Fu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
120
|
Yin S, Liu J, Kang Y, Lin Y, Li D, Shao L. Interactions of nanomaterials with ion channels and related mechanisms. Br J Pharmacol 2019; 176:3754-3774. [PMID: 31290152 DOI: 10.1111/bph.14792] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 06/10/2019] [Accepted: 07/01/2019] [Indexed: 12/15/2022] Open
Abstract
The pharmacological potential of nanotechnology, especially in drug delivery and bioengineering, has developed rapidly in recent decades. Ion channels, which are easily targeted by external agents, such as nanomaterials (NMs) and synthetic drugs, due to their unique structures, have attracted increasing attention in the fields of nanotechnology and pharmacology for the treatment of ion channel-related diseases. NMs have significant effects on ion channels, and these effects are manifested in many ways, including changes in ion currents, kinetic characteristics and channel distribution. Subsequently, intracellular ion homeostasis, signalling pathways, and intracellular ion stores are affected, leading to the initiation of a range of biological processes. However, the effect of the interactions of NMs with ion channels is an interesting topic that remains obscure. In this review, we have summarized the recent research progress on the direct and indirect interactions between NMs and ion channels and discussed the related molecular mechanisms, which are crucial to the further development of ion channel-related nanotechnological applications.
Collapse
Affiliation(s)
- Suhan Yin
- Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Jia Liu
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyuan Kang
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuqing Lin
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dongjian Li
- Liwan District Stomatology Hospital, Guangzhou, China
| | - Longquan Shao
- Nanfang Hospital, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
121
|
Huang KF, Ma KH, Jhap TY, Liu PS, Chueh SH. Ultraviolet B irradiation induced Nrf2 degradation occurs via activation of TRPV1 channels in human dermal fibroblasts. Free Radic Biol Med 2019; 141:220-232. [PMID: 31220549 DOI: 10.1016/j.freeradbiomed.2019.06.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 06/10/2019] [Accepted: 06/15/2019] [Indexed: 12/21/2022]
Abstract
Ultraviolet (UV) irradiation causes cellular oxidative stress. Under redox imbalance, Keap1-dependent Nrf2 degradation is minimal. In this study, we examined the role of Ca2+ in Nrf2 homeostasis after UVB irradiation using human dermal fibroblasts. UVB irradiation stimulates 12-lipoxygenase and the product 12-hydroxyeicosatetraenoic acid then activates TRPV1 increasing the cell's cytosolic Ca2+ concentration. UVB irradiation induced reactive oxygen species generation and apoptosis are inhibited in the absence of Ca2+ or in the presence of either a 12-lipoxygenase inhibitor or a TRPV1 inhibitor during and after UVB irradiation. Thus, the Ca2+ increase via TRPV1 is a critical factor in UVB irradiation induced oxidative stress. UVB irradiation induces a Ca2+ dependent Nrf2 degradation and thus activation of TRPV1 with 12-hydroxyeicosatetraenoic acid also decreasing Nrf2 levels. UVB irradiation induced Nrf2 degradation is inhibited by co-treatment of cells with W-7, cyclosporin A, SB-216763 or MG-132, which are inhibitors of calmodulin, calcineurin, GSK3β and the proteasome, respectively. Furthermore, UVB irradiation in parallel induces GSK3β dephosphorylation in a Ca2+ dependent manner. Co-immunoprecipitation showed that UVB irradiation induces an increase in Nrf2 phosphorylation, an increase in the binding of β-TrCP and Nrf2, and an increase in Nrf2 ubiquitination; these effects are all Ca2+ dependent. These findings suggest that UVB irradiation induced GSK3β activation in a Ca2+ dependent manner, which then stimulates the phosphorylation and ubiquitination of Nrf2 via β-TrCP. Indeed, silencing of β-TrCP was found to inhibit UVB irradiation-induced oxidative stress, Nrf2 degradation and apoptosis, while it had no effect on the Ca2+ increase. Taken together, our results suggest that a Ca2+ influx via TRPV1 is responsible for UVB irradiation-induced Nrf2 degradation and that modulation of the Ca2+-calmodulin-calcineurin-GSK3β-Nrf2-β-TrCP-Cullin-1 pathway may explain Ca2+ dependent Nrf2 degradation.
Collapse
Affiliation(s)
- Kuo-Feng Huang
- Division of Plastic Surgery, Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan, ROC
| | - Kuo-Hsing Ma
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Tian-You Jhap
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Pei-Shan Liu
- Department of Microbiology, Soochow University, Taipei, Taiwan, ROC
| | - Sheau-Huei Chueh
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, ROC.
| |
Collapse
|
122
|
Zhang X, Gibhardt CS, Will T, Stanisz H, Körbel C, Mitkovski M, Stejerean I, Cappello S, Pacheu‐Grau D, Dudek J, Tahbaz N, Mina L, Simmen T, Laschke MW, Menger MD, Schön MP, Helms V, Niemeyer BA, Rehling P, Vultur A, Bogeski I. Redox signals at the ER-mitochondria interface control melanoma progression. EMBO J 2019; 38:e100871. [PMID: 31304984 PMCID: PMC6669928 DOI: 10.15252/embj.2018100871] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 05/21/2019] [Accepted: 05/23/2019] [Indexed: 12/20/2022] Open
Abstract
Reactive oxygen species (ROS) are emerging as important regulators of cancer growth and metastatic spread. However, how cells integrate redox signals to affect cancer progression is not fully understood. Mitochondria are cellular redox hubs, which are highly regulated by interactions with neighboring organelles. Here, we investigated how ROS at the endoplasmic reticulum (ER)-mitochondria interface are generated and translated to affect melanoma outcome. We show that TMX1 and TMX3 oxidoreductases, which promote ER-mitochondria communication, are upregulated in melanoma cells and patient samples. TMX knockdown altered mitochondrial organization, enhanced bioenergetics, and elevated mitochondrial- and NOX4-derived ROS. The TMX-knockdown-induced oxidative stress suppressed melanoma proliferation, migration, and xenograft tumor growth by inhibiting NFAT1. Furthermore, we identified NFAT1-positive and NFAT1-negative melanoma subgroups, wherein NFAT1 expression correlates with melanoma stage and metastatic potential. Integrative bioinformatics revealed that genes coding for mitochondrial- and redox-related proteins are under NFAT1 control and indicated that TMX1, TMX3, and NFAT1 are associated with poor disease outcome. Our study unravels a novel redox-controlled ER-mitochondria-NFAT1 signaling loop that regulates melanoma pathobiology and provides biomarkers indicative of aggressive disease.
Collapse
Affiliation(s)
- Xin Zhang
- Molecular PhysiologyInstitute of Cardiovascular PhysiologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
- BiophysicsCIPMMSaarland UniversityHomburgGermany
| | - Christine S Gibhardt
- Molecular PhysiologyInstitute of Cardiovascular PhysiologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Thorsten Will
- Center for BioinformaticsSaarland UniversitySaarbrückenGermany
| | - Hedwig Stanisz
- Department of Dermatology, Venereology and AllergologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Christina Körbel
- Institute for Clinical and Experimental SurgerySaarland UniversityHomburgGermany
| | - Miso Mitkovski
- Light Microscopy FacilityMax Planck Institute for Experimental MedicineGöttingenGermany
| | - Ioana Stejerean
- Molecular PhysiologyInstitute of Cardiovascular PhysiologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Sabrina Cappello
- Molecular PhysiologyInstitute of Cardiovascular PhysiologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - David Pacheu‐Grau
- Department of Cellular BiochemistryUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Jan Dudek
- Department of Cellular BiochemistryUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Nasser Tahbaz
- Department of Cell BiologyUniversity of AlbertaEdmontonABCanada
| | - Lucas Mina
- Department of Cell BiologyUniversity of AlbertaEdmontonABCanada
| | - Thomas Simmen
- Department of Cell BiologyUniversity of AlbertaEdmontonABCanada
| | - Matthias W Laschke
- Institute for Clinical and Experimental SurgerySaarland UniversityHomburgGermany
| | - Michael D Menger
- Institute for Clinical and Experimental SurgerySaarland UniversityHomburgGermany
| | - Michael P Schön
- Department of Dermatology, Venereology and AllergologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Volkhard Helms
- Center for BioinformaticsSaarland UniversitySaarbrückenGermany
| | | | - Peter Rehling
- Department of Cellular BiochemistryUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
- Max Planck Institute for Biophysical ChemistryGöttingenGermany
| | - Adina Vultur
- Molecular PhysiologyInstitute of Cardiovascular PhysiologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
| | - Ivan Bogeski
- Molecular PhysiologyInstitute of Cardiovascular PhysiologyUniversity Medical CenterGeorg‐August‐UniversityGöttingenGermany
- BiophysicsCIPMMSaarland UniversityHomburgGermany
| |
Collapse
|
123
|
Lee JR, Lee JY, Kim HJ, Hahn MJ, Kang JS, Cho H. The inhibition of chloride intracellular channel 1 enhances Ca 2+ and reactive oxygen species signaling in A549 human lung cancer cells. Exp Mol Med 2019; 51:1-11. [PMID: 31316050 PMCID: PMC6802611 DOI: 10.1038/s12276-019-0279-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 02/22/2019] [Accepted: 03/18/2019] [Indexed: 01/22/2023] Open
Abstract
Chloride intracellular channel 1 (CLIC1) is a promising therapeutic target in cancer due to its intrinsic characteristics; it is overexpressed in specific tumor types and its localization changes from cytosolic to surface membrane depending on activities and cell cycle progression. Ca2+ and reactive oxygen species (ROS) are critical signaling molecules that modulate diverse cellular functions, including cell death. In this study, we investigated the function of CLIC1 in Ca2+ and ROS signaling in A549 human lung cancer cells. Depletion of CLIC1 via shRNAs in A549 cells increased DNA double-strand breaks both under control conditions and under treatment with the putative anticancer agent chelerythrine, accompanied by a concomitant increase in the p-JNK level. CLIC1 knockdown greatly increased basal ROS levels, an effect prevented by BAPTA-AM, an intracellular calcium chelator. Intracellular Ca2+ measurements clearly showed that CLIC1 knockdown significantly increased chelerythrine-induced Ca2+ signaling as well as the basal Ca2+ level in A549 cells compared to these levels in control cells. Suppression of extracellular Ca2+ restored the basal Ca2+ level in CLIC1-knockdown A549 cells relative to that in control cells, implying that CLIC1 regulates [Ca2+]i through Ca2+ entry across the plasma membrane. Consistent with this finding, the L-type Ca2+ channel (LTCC) blocker nifedipine reduced the basal Ca2+ level in CLIC1 knockdown cells to that in control cells. Taken together, our results demonstrate that CLIC1 knockdown induces an increase in the intracellular Ca2+ level via LTCC, which then triggers excessive ROS production and consequent JNK activation. Thus, CLIC1 is a key regulator of Ca2+ signaling in the control of cancer cell survival.
Collapse
Affiliation(s)
- Jae-Rin Lee
- 0000 0001 2181 989Xgrid.264381.aDepartment of Molecular Cell Biology, Sungkyunkwan University, Suwon, Korea ,0000 0001 2181 989Xgrid.264381.aSingle Cell Network Research Center, Sungkyunkwan University, Suwon, Korea
| | - Jong-Yoon Lee
- 0000 0001 2181 989Xgrid.264381.aSingle Cell Network Research Center, Sungkyunkwan University, Suwon, Korea ,0000 0001 2181 989Xgrid.264381.aDepartment of Physiology, Sungkyunkwan University, Suwon, Korea
| | - Hyun-Ji Kim
- 0000 0001 2181 989Xgrid.264381.aSingle Cell Network Research Center, Sungkyunkwan University, Suwon, Korea ,0000 0001 2181 989Xgrid.264381.aDepartment of Physiology, Sungkyunkwan University, Suwon, Korea
| | - Myong-Joon Hahn
- 0000 0001 2181 989Xgrid.264381.aDepartment of Molecular Cell Biology, Sungkyunkwan University, Suwon, Korea
| | - Jong-Sun Kang
- 0000 0001 2181 989Xgrid.264381.aDepartment of Molecular Cell Biology, Sungkyunkwan University, Suwon, Korea ,0000 0001 2181 989Xgrid.264381.aSingle Cell Network Research Center, Sungkyunkwan University, Suwon, Korea
| | - Hana Cho
- 0000 0001 2181 989Xgrid.264381.aSingle Cell Network Research Center, Sungkyunkwan University, Suwon, Korea ,0000 0001 2181 989Xgrid.264381.aDepartment of Physiology, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
124
|
Zeferino RC, Mota NSRS, Grinevicius VMAS, Filipe KB, Sulis PM, Silva FRMB, Filho DW, Pich CT, Pedrosa RC. Targeting ROS overgeneration by N-benzyl-2-nitro-1-imidazole-acetamide as a potential therapeutic reposition approach for cancer therapy. Invest New Drugs 2019; 38:785-799. [PMID: 31257554 DOI: 10.1007/s10637-019-00820-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 06/17/2019] [Indexed: 11/25/2022]
Abstract
BackgroundWe investigated the role of reactive oxygen species (ROS) in the anticancer mechanism of N-benzyl-2-nitro-1-imidazole-acetamide (BZN), a drug used in Chagas' disease treatment. MethodsBALB/c mice, inoculated with Ehrlich ascites carcinoma (EAC), were treated with BZN or BZN + Nacylcysteine (NAC) or NAC for 9 days. Subsequently, the inhibition of tumor growth and angiogenesis as well as animal survival were evaluated. Apoptosis and the cell cycle were evaluated using fluorescence microscopy and flow cytometry, while oxidative stress was evaluated by measuring TBARS content, DNA damage, calcium influx and ROS generation and antioxidant defenses (CAT, SOD, GPx, GST and GR). Immunoblotting was used to evaluate key death and cell cycle proteins. Results BZN treatment inhibited tumor progression (79%), angiogenesis (2.8-fold) and increased animal survival (29%). Moreover, BZN increased ROS levels (42%), calcium influx (55%), TBARS contents (1.9-fold), SOD (4.4-fold), GPx (17.5-fold) and GST (3-fold) activities and GSH depletion (2.5-fold) also caused DNA fragmentation (7.6-fold), increased cleaved PARP and promoted the trapping of cells in the G1 phase, as corroborated by the reduction in cyclin A and increased CDK2 protein levels. In silico DNA and molecular dynamic simulations showed H-bonds and hydrophobic interactions that were confirmed by circular dichroism. Increased apoptosis (232%), induced by treatment with BZN, was demonstrated by apoptotic cell staining and p53 level. Conclusion The current findings indicate that BZN acts as a tumor growth inhibitor and anti-angiogenic agent by ROS overgeneration, which interact with DNA causing damage and triggering apoptosis.
Collapse
Affiliation(s)
- Rodrigo C Zeferino
- Biochemistry Department, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Nádia S R S Mota
- Biochemistry Department, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | | | - Karina B Filipe
- Department of Clinical Analysis, Federal University of Paraná (UFPR), Curitiba, PR, Brazil
| | - Paola M Sulis
- Biochemistry Department, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Fátima R M B Silva
- Biochemistry Department, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Danilo W Filho
- Ecology and Zoology Department, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Claus T Pich
- Center for Sciences, Technologies and Health, Federal University of Santa Catarina (UFSC), Araranguá, SC, Brazil
| | - Rozangela C Pedrosa
- Biochemistry Department, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil.
| |
Collapse
|
125
|
ROS-Mediated Cancer Cell Killing through Dietary Phytochemicals. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9051542. [PMID: 31217841 PMCID: PMC6536988 DOI: 10.1155/2019/9051542] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/18/2019] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) promote carcinogenesis by inducing genetic mutations, activating oncogenes, and raising oxidative stress, which all influence cell proliferation, survival, and apoptosis. Cancer cells display redox imbalance due to increased ROS level compared to normal cells. This unique feature in cancer cells may, therefore, be exploited for targeted therapy. Over the past few decades, natural compounds have attracted attention as potential cancer therapies because of their ability to maintain cellular redox homeostasis with minimal toxicity. Preclinical studies show that bioactive dietary polyphenols exert antitumor effects by inducing ROS-mediated cytotoxicity in cancer cells. These bioactive compounds also regulate cell proliferation, survival, and apoptotic and antiapoptotic signalling pathways. In this review, we discuss (i) how ROS is generated and (ii) regulated and (iii) the cell signalling pathways affected by ROS. We also discuss (iv) the various dietary phytochemicals that have been implicated to have cancer therapeutic effects through their ROS-related functions.
Collapse
|
126
|
Gu J, Wang Y, Liu Y, Shi M, Yin L, Hou Y, Zhou Y, Chu Wong CK, Chen D, Guo Z, Shi H. Inhibition of Autophagy Alleviates Cadmium-Induced Mouse Spleen and Human B Cells Apoptosis. Toxicol Sci 2019; 170:109-122. [DOI: 10.1093/toxsci/kfz089] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Abstract
Cadmium (Cd) is a toxic heavy metal that can accumulate and cause severe damage to many organs, such as liver, kidney, lung, etc. Cd also significantly suppresses immunity, however, the underlying mechanism involved in Cd-induced immunnotoxicity is still unclear. The present study indicated that semichronic Cd exposure (7 days) induced apoptotic damage of mouse spleen. In human Ramos B cells, Cd exposure also induced apoptosis, which was dependent on Cd-induced vacuole membrane protein 1 (VMP1) expression and autophagy. Cd-induced autophagy and apoptosis were abated when VMP1 expression was knockdown. In addition, Cd-induced VMP1 expression, autophagy, and apoptosis were dependent on the elevation of Ca2+ and reactive oxygen species (ROS). More important, Cd exposure also induced VMP1 expression and autophagy in mouse spleen tissue, and the intraperitoneal injection of the autophagy inhibitor chloroquine (CQ) into mice effectively reduced Cd-induced spleen apoptotic damage. Taken together, these results indicate Cd-induced autophagy, promotes apoptosis in immune cells, and inhibition of autophagy can alleviate Cd-induced spleen and immune cell apoptosis. This study might provide the groundwork for future studies on Cd-induced immunomodulatory effects and immune diseases.
Collapse
Affiliation(s)
- Jie Gu
- Institute of Life Sciences, Jiangsu University
| | - Yanwei Wang
- Institute of Life Sciences, Jiangsu University
| | - Yanmin Liu
- Institute of Life Sciences, Jiangsu University
| | - Meilin Shi
- Institute of Life Sciences, Jiangsu University
| | - Liangdong Yin
- Department of Osteology, The Third Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, China
| | | | - Yang Zhou
- Institute of Life Sciences, Jiangsu University
| | | | - Dongfeng Chen
- Institute of Life Sciences, Jiangsu University
- Department of Rheumatology and Inflammation Research, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu 210023, China
| | - Haifeng Shi
- Institute of Life Sciences, Jiangsu University
| |
Collapse
|
127
|
Store-Operated Calcium Entry Contributes to Cisplatin-Induced Cell Death in Non-Small Cell Lung Carcinoma. Cancers (Basel) 2019; 11:cancers11030430. [PMID: 30917547 PMCID: PMC6468672 DOI: 10.3390/cancers11030430] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/19/2019] [Accepted: 03/22/2019] [Indexed: 12/11/2022] Open
Abstract
Cisplatin (CDDP) is one of the principal chemotherapeutic agents used for the first-line treatment of many malignancies, including non-small cell lung carcinoma (NSCLC). Despite its use for over 40 years, its mechanism of action is not yet fully understood. Store-operated calcium entry (SOCE), the main pathway allowing Ca2+ entry in non-excitable cells, is involved in tumorogenesis, cancer progression and chemoresistance. It has become an attractive target in cancer treatment. In this study, we showed that siRNA-mediated depletion of stromal interaction molecule 1 (STIM1) and transient receptor potential channel 1 (TRPC1), two players of the store-operated calcium entry, dramatically reduced CDDP cytotoxicity in NSCLC cells. This was associated with an inhibition of the DNA damage response (DDR) triggered by CDDP. Moreover, STIM1 depletion also reduced CDDP-dependent oxidative stress. In parallel, SOCE activation induced Ca2+ entry into the mitochondria, a major source of reactive oxygen species (ROS) within the cell. This effect was highly decreased in STIM1-depleted cells. We then conclude that mitochondrial Ca2+ peak associated to the SOCE contributes to CDDP-induced ROS production, DDR and subsequent apoptosis. To the best of our knowledge, this is the first time that it is shown that Ca2+ signalling constitutes an initial step in CDDP-induced apoptosis.
Collapse
|
128
|
Huang K, Ma K, Chang Y, Lo L, Jhap T, Su Y, Liu P, Chueh S. Baicalein inhibits matrix metalloproteinase 1 expression via activation of
TRPV
1‐Ca‐
ERK
pathway in ultraviolet B–irradiated human dermal fibroblasts. Exp Dermatol 2019; 28:568-575. [DOI: 10.1111/exd.13912] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/23/2019] [Accepted: 02/18/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Kuo‐Feng Huang
- Division of Plastic Surgery Department of Surgery Chi Mei Medical Center Tainan Taiwan Republic of China
| | - Kuo‐Hsing Ma
- Department of Biology and Anatomy National Defense Medical Center Taipei Taiwan Republic of China
| | - Yen‐Jung Chang
- Department of Biochemistry National Defense Medical Center Taipei Taiwan Republic of China
| | - Liang‐Chuan Lo
- Department of Biochemistry National Defense Medical Center Taipei Taiwan Republic of China
| | - Tian‐You Jhap
- Department of Biochemistry National Defense Medical Center Taipei Taiwan Republic of China
| | - Yu‐Hua Su
- Department of Biochemistry National Defense Medical Center Taipei Taiwan Republic of China
| | - Pei‐Shan Liu
- Department of Microbiology Soochow University Taipei Taiwan Republic of China
| | - Sheau‐Huei Chueh
- Department of Biochemistry National Defense Medical Center Taipei Taiwan Republic of China
| |
Collapse
|
129
|
Lepetsos P, Papavassiliou KA, Papavassiliou AG. Redox and NF-κB signaling in osteoarthritis. Free Radic Biol Med 2019; 132:90-100. [PMID: 30236789 DOI: 10.1016/j.freeradbiomed.2018.09.025] [Citation(s) in RCA: 247] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 09/12/2018] [Accepted: 09/16/2018] [Indexed: 02/07/2023]
Abstract
Human cells have to deal with the constant production of reactive oxygen species (ROS). Although ROS overproduction might be harmful to cell biology, there are plenty of data showing that moderate levels of ROS control gene expression by maintaining redox signaling. Osteoarthritis (OA) is the most common joint disorder with a multi-factorial etiology including overproduction of ROS. ROS overproduction in OA modifies intracellular signaling, chondrocyte life cycle, metabolism of cartilage matrix and contributes to synovial inflammation and dysfunction of the subchondral bone. In arthritic tissues, the NF-κB signaling pathway can be activated by pro-inflammatory cytokines, mechanical stress, and extracellular matrix degradation products. This activation results in regulation of expression of many cytokines, inflammatory mediators, transcription factors, and several matrix-degrading enzymes. Overall, NF-κB signaling affects cartilage matrix remodeling, chondrocyte apoptosis, synovial inflammation, and has indirect stimulatory effects on downstream regulators of terminal chondrocyte differentiation. Interaction between redox signaling and NF-κB transcription factors seems to play a distinctive role in OA pathogenesis.
Collapse
Affiliation(s)
- Panagiotis Lepetsos
- Fourth Department of Orthopaedics & Trauma, 'KAT' General Hospital, Kifissia, 14561 Athens, Greece
| | - Kostas A Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street, 11527 Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street, 11527 Athens, Greece.
| |
Collapse
|
130
|
Nagar M, Tilvawala R, Thompson PR. Thioredoxin Modulates Protein Arginine Deiminase 4 (PAD4)-Catalyzed Citrullination. Front Immunol 2019; 10:244. [PMID: 30853960 PMCID: PMC6396667 DOI: 10.3389/fimmu.2019.00244] [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: 07/09/2018] [Accepted: 01/28/2019] [Indexed: 01/01/2023] Open
Abstract
Protein citrullination is a post-translational modification catalyzed by the protein arginine deiminases (PADs). This modification plays a crucial role in the pathophysiology of numerous autoimmune disorders including RA. Recently, there has been a growing interest in investigating physiological regulators of PAD activity to understand the primary cause of the associated disorders. Apart from calcium, it is well-documented that a reducing environment activates the PADs. Although the concentration of thioredoxin (hTRX), an oxidoreductase that maintains the cellular reducing environment, is elevated in RA patients, its contribution toward RA progression or PAD activity has not been explored. Herein, we demonstrate that hTRX activates PAD4. Kinetic characterization of PAD4 using hTRX as the reducing agent yielded parameters that are comparable to those obtained with a routinely used non-physiological reducing agent, e.g., DTT, suggesting the importance of hTRX in PAD regulation under physiological conditions. Furthermore, we show that various hTRX mutants, including redox inactive hTRX variants, are capable of activating PAD4. This indicates a mechanism that does not require oxidoreductase activity. Indeed, we observed non-covalent interactions between PAD4 and hTRX variants, and propose that these redox-independent interactions are sufficient for hTRX-mediated PAD4 activation.
Collapse
Affiliation(s)
- Mitesh Nagar
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, United States.,Program in Chemical Biology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Ronak Tilvawala
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, United States.,Program in Chemical Biology, University of Massachusetts Medical School, Worcester, MA, United States
| | - Paul R Thompson
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, United States.,Program in Chemical Biology, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
131
|
Parascandolo A, Laukkanen MO. Carcinogenesis and Reactive Oxygen Species Signaling: Interaction of the NADPH Oxidase NOX1-5 and Superoxide Dismutase 1-3 Signal Transduction Pathways. Antioxid Redox Signal 2019; 30:443-486. [PMID: 29478325 PMCID: PMC6393772 DOI: 10.1089/ars.2017.7268] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Reduction/oxidation (redox) balance could be defined as an even distribution of reduction and oxidation complementary processes and their reaction end products. There is a consensus that aberrant levels of reactive oxygen species (ROS), commonly observed in cancer, stimulate primary cell immortalization and progression of carcinogenesis. However, the mechanism how different ROS regulate redox balance is not completely understood. Recent Advances: In the current review, we have summarized the main signaling cascades inducing NADPH oxidase NOX1-5 and superoxide dismutase (SOD) 1-3 expression and their connection to cell proliferation, immortalization, transformation, and CD34+ cell differentiation in thyroid, colon, lung, breast, and hematological cancers. CRITICAL ISSUES Interestingly, many of the signaling pathways activating redox enzymes or mediating the effect of ROS are common, such as pathways initiated from G protein-coupled receptors and tyrosine kinase receptors involving protein kinase A, phospholipase C, calcium, and small GTPase signaling molecules. FUTURE DIRECTIONS The clarification of interaction of signal transduction pathways could explain how cells regulate redox balance and may even provide means to inhibit the accumulation of harmful levels of ROS in human pathologies.
Collapse
|
132
|
Kappel S, Borgström A, Stokłosa P, Dörr K, Peinelt C. Store-operated calcium entry in disease: Beyond STIM/Orai expression levels. Semin Cell Dev Biol 2019; 94:66-73. [PMID: 30630032 DOI: 10.1016/j.semcdb.2019.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/29/2018] [Accepted: 01/05/2019] [Indexed: 12/19/2022]
Abstract
Precise intracellular calcium signaling is crucial to numerous cellular functions. In non-excitable cells, store-operated calcium entry (SOCE) is a key step in the generation of intracellular calcium signals. Tight regulation of SOCE is important, and dysregulation is involved in several pathophysiological cellular malfunctions. The current underlying SOCE, calcium release-activated calcium current (ICRAC), was first discovered almost three decades ago. Since its discovery, the molecular components of ICRAC, Orai1 and stromal interaction molecule 1 (STIM1), have been extensively investigated. Several regulatory mechanisms and proteins contribute to alterations in SOCE and cellular malfunctions in cancer, immune and neurodegenerative diseases, inflammation, and neuronal disorders. This review summarizes these regulatory mechanisms, including glycosylation, pH sensing, and the regulatory proteins golli, α-SNAP, SARAF, ORMDL3, CRACR2A, and TRPM4 channels.
Collapse
Affiliation(s)
- Sven Kappel
- Institute of Biochemistry and Molecular Medicine, National Center of Competence in Research NCCR TransCure, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland
| | - Anna Borgström
- Institute of Biochemistry and Molecular Medicine, National Center of Competence in Research NCCR TransCure, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland
| | - Paulina Stokłosa
- Institute of Biochemistry and Molecular Medicine, National Center of Competence in Research NCCR TransCure, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland
| | | | - Christine Peinelt
- Institute of Biochemistry and Molecular Medicine, National Center of Competence in Research NCCR TransCure, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland.
| |
Collapse
|
133
|
Paroxetine Induces Apoptosis of Human Breast Cancer MCF-7 Cells through Ca 2+-and p38 MAP Kinase-Dependent ROS Generation. Cancers (Basel) 2019; 11:cancers11010064. [PMID: 30634506 PMCID: PMC6356564 DOI: 10.3390/cancers11010064] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 01/04/2019] [Indexed: 12/16/2022] Open
Abstract
Depression is more common in women with breast cancer than the general population. Selective serotonin reuptake inhibitors (SSRIs), a group of antidepressants, are widely used for the treatment of patients with depression and a range of anxiety-related disorders. The association between the use of antidepressant medication and breast cancer is controversial. In this study, we investigated whether and how SSRIs induce the death of human breast cancer MCF-7 cells. Of the antidepressants tested in this study (amitriptyline, bupropion, fluoxetine, paroxetine, and tianeptine), paroxetine most reduced the viability of MCF-7 cells in a time-and dose-dependent manner. The exposure of MCF-7 cells to paroxetine resulted in mitochondrion-mediated apoptosis, which is assessed by increase in the number of cells with sub-G1 DNA content, caspase-8/9 activation, poly (ADP-ribose) polymerase cleavage, and Bax/Bcl-2 ratio and a reduction in the mitochondrial membrane potential. Paroxetine increased a generation of reactive oxygen species (ROS), intracellular Ca2+ levels, and p38 MAPK activation. The paroxetine-induced apoptotic events were reduced by ROS scavengers and p38 MAPK inhibitor, and the paroxetine’s effect was dependent on extracellular Ca2+ level. Paroxetine also showed a synergistic effect on cell death induced by chemotherapeutic drugs in MCF-7 and MDA-MB-231 cells. Our results showed that paroxetine induced apoptosis of human breast cancer MCF-7 cells through extracellular Ca2+-and p38 MAPK-dependent ROS generation. These results suggest that paroxetine may serve as an anticancer adjuvant to current cancer therapies for breast cancer patients with or without depression.
Collapse
|
134
|
Gibhardt CS, Vultur A, Bogeski I. Measuring Calcium and ROS by Genetically Encoded Protein Sensors and Fluorescent Dyes. Methods Mol Biol 2019; 1925:183-196. [PMID: 30674028 DOI: 10.1007/978-1-4939-9018-4_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Oxidative modifications of cellular building blocks such as proteins, lipids, and DNA have a major impact on cell behavior, fate, and clinical outcome. Reactive oxygen species (ROS) are important factors that influence these redox processes. Calcium ion (Ca2+) dynamics and signals are also essential regulators of key cellular processes. Therefore, the combined and precise monitoring of ROS and Ca2+ in single cells, with a high spatial and temporal resolution and in physiological environments, is essential to better understand their functional impact. Here, we describe protocols to detect one of the most prominent ROS (hydrogen peroxide, H2O2) using genetically encoded protein sensors and fluorescent dyes. We also provide guidelines on how to simultaneously detect Ca2+ and H2O2 and how to examine the influence of Ca2+ signals on cellular ROS production and vice versa.
Collapse
Affiliation(s)
- Christine S Gibhardt
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Adina Vultur
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Ivan Bogeski
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany.
| |
Collapse
|
135
|
Yang J. The role of reactive oxygen species in angiogenesis and preventing tissue injury after brain ischemia. Microvasc Res 2018; 123:62-67. [PMID: 30594490 DOI: 10.1016/j.mvr.2018.12.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/24/2018] [Accepted: 12/26/2018] [Indexed: 02/06/2023]
Abstract
Oxidative stress, which is defined as an imbalance between proxidant and antioxidant systems, is the essential mechanism involving in the ischemic process. During the early stage of brain ischemia, reactive oxygen species (ROS) are increased. Increased ROS are thought of a consequence of brain ischemia and exacerbating disease due to inducing cell death, apoptosis and senescence by oxidative stress. During brain tissue repair, ROS are act as signaling molecules and may be benefical for regulating angiogenesis and preventing tissue injury. New blood vessel formation is essentially required for rescuing tissue from brain ischemia. In ischemic conditions, ROS promotes angiogenesis, either directly or via the generation of active oxidation products. ROS-induced angiogenesis involves several signaling pathways. This paper reviewed current understanding of the role of ROS as a mediator and modulator of angiogenesis in brain ischemia.
Collapse
Affiliation(s)
- Jiping Yang
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, 215 West Heping Road, Shijiazhuang 050000, Hebei Province, China.
| |
Collapse
|
136
|
Wang Y, Luo YH, Piao XJ, Shen GN, Meng LQ, Zhang Y, Wang JR, Li JQ, Wang H, Xu WT, Liu Y, Zhang Y, Zhang T, Wang SN, Sun HN, Han YH, Jin MH, Zang YQ, Zhang DJ, Jin CH. Novel 1,4‑naphthoquinone derivatives induce reactive oxygen species‑mediated apoptosis in liver cancer cells. Mol Med Rep 2018; 19:1654-1664. [PMID: 30592276 PMCID: PMC6390020 DOI: 10.3892/mmr.2018.9785] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 11/15/2018] [Indexed: 12/13/2022] Open
Abstract
Derivatives of 1,4-naphthoquinone have excellent anti-cancer effects, but their use has been greatly limited due to their serious side effects. To develop compounds with decreased side effects and improved anti-cancer activity, two novel types of 1,4-naphthoquinone derivatives, 2,3-dihydro-2,3-epoxy-2-propylsulfonyl-5,8-dimethoxy-1,4-naphthoquinone (EPDMNQ) and 2,3-dihydro-2,3-epoxy-2-nonylsulfonyl-5,8-dimethoxy-1,4-naphthoquinone (ENDMNQ) were synthesized and their anti-tumor activities were investigated. The effects of EPDMNQ and ENDMNQ on cell viability, apoptosis and accumulation of reactive oxygen species (ROS) in liver cancer cells were determined by MTT cell viability assay and flow cytometry. The expression levels of mitochondrial, mitogen activated protein kinase (MAPK) and signal transducer and activator of transcription 3 (STAT3) signaling pathway-associated proteins in Hep3B liver cancer cells were analyzed by western blot analysis. The results demonstrated that EPDMNQ and ENDMNQ inhibited the proliferation of liver cancer Hep3B, HepG2, and Huh7 cell lines but not that of normal liver L-02, normal lung IMR-90 and stomach GES-1 cell lines. The number of apoptotic cells and ROS levels were significantly increased following treatment with EPDMNQ and ENDMNQ, and these effects were blocked by the ROS inhibitor N-acetyl-L-cysteine (NAC) in Hep3B cells. EPDMNQ and ENDMNQ induced apoptosis by upregulating the protein expression of p38 MAPK and c-Jun N-terminal kinase and downregulating extracellular signal-regulated kinase and STAT3; these effects were inhibited by NAC. The results of the present study demonstrated that EPDMNQ and ENDMNQ induced apoptosis through ROS-modulated MAPK and STAT3 signaling pathways in Hep3B cells. Therefore, these novel 1,4-naphthoquinone derivatives may be useful as anticancer agents for the treatment of liver cancer.
Collapse
Affiliation(s)
- Yue Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Ying-Hua Luo
- Department of Grass Science, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Xian-Ji Piao
- Department of Gynaecology and Obstetrics, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, Heilongjiang 163316, P.R. China
| | - Gui-Nan Shen
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Ling-Qi Meng
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yi Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Jia-Ru Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Jin-Qian Li
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Hao Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Wan-Ting Xu
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yang Liu
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yu Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Tong Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Shi-Nong Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Hu-Nan Sun
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Ying-Hao Han
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Mei-Hua Jin
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yan-Qing Zang
- Department of Food Science and Engineering, College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Dong-Jie Zhang
- Department of Food Science and Engineering, College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Cheng-Hao Jin
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| |
Collapse
|
137
|
Jaquenod De Giusti C, Roman B, Das S. The Influence of MicroRNAs on Mitochondrial Calcium. Front Physiol 2018; 9:1291. [PMID: 30298016 PMCID: PMC6160583 DOI: 10.3389/fphys.2018.01291] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/27/2018] [Indexed: 01/13/2023] Open
Abstract
Abnormal mitochondrial calcium ([Ca2+]m) handling and energy deficiency results in cellular dysfunction and cell death. Recent studies suggest that nuclear-encoded microRNAs (miRNA) are able to translocate in to the mitochondrial compartment, and modulate mitochondrial activities, including [Ca2+]m uptake. Apart from this subset of miRNAs, there are several miRNAs that have been reported to target genes that play a role in maintaining [Ca2+]m levels in the cytoplasm. It is imperative to validate miRNAs that alter [Ca2+]m handling, and thereby alter cellular fate. The focus of this review is to highlight the mitochondrial miRNAs (MitomiRs), and other cytosolic miRNAs that target mRNAs which play an important role in [Ca2+]m handling.
Collapse
Affiliation(s)
- Carolina Jaquenod De Giusti
- Centro de Investigaciones Cardiovasculares CIC-CONICET, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Barbara Roman
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Samarjit Das
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
138
|
Kalinowska M, Mazur L, Jabłońska-Trypuć A, Lewandowski W. A new calcium 2,5-dihydroxybenzoate: Synthesis, characterization and antioxidant studies and stress mediated cytotoxity in MCF-7 cells. JOURNAL OF SAUDI CHEMICAL SOCIETY 2018. [DOI: 10.1016/j.jscs.2017.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
139
|
Prieto-Bermejo R, Romo-González M, Pérez-Fernández A, Ijurko C, Hernández-Hernández Á. Reactive oxygen species in haematopoiesis: leukaemic cells take a walk on the wild side. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:125. [PMID: 29940987 PMCID: PMC6019308 DOI: 10.1186/s13046-018-0797-0] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/15/2018] [Indexed: 02/08/2023]
Abstract
Oxidative stress is related to ageing and degenerative diseases, including cancer. However, a moderate amount of reactive oxygen species (ROS) is required for the regulation of cellular signalling and gene expression. A low level of ROS is important for maintaining quiescence and the differentiation potential of haematopoietic stem cells (HSCs), whereas the level of ROS increases during haematopoietic differentiation; thus, suggesting the importance of redox signalling in haematopoiesis. Here, we will analyse the importance of ROS for haematopoiesis and include evidence showing that cells from leukaemia patients live under oxidative stress. The potential sources of ROS will be described. Finally, the level of oxidative stress in leukaemic cells can also be harnessed for therapeutic purposes. In this regard, the reliance of front-line anti-leukaemia chemotherapeutics on increased levels of ROS for their mechanism of action, as well as the active search for novel compounds that modulate the redox state of leukaemic cells, will be analysed.
Collapse
Affiliation(s)
- Rodrigo Prieto-Bermejo
- Department of Biochemistry and Molecular Biology, University of Salamanca, Lab. 122, Edificio Departamental, Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain.,IBSAL (Instituto de investigación Biomédica de Salamanca), Salamanca, Spain
| | - Marta Romo-González
- Department of Biochemistry and Molecular Biology, University of Salamanca, Lab. 122, Edificio Departamental, Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain.,IBSAL (Instituto de investigación Biomédica de Salamanca), Salamanca, Spain
| | - Alejandro Pérez-Fernández
- Department of Biochemistry and Molecular Biology, University of Salamanca, Lab. 122, Edificio Departamental, Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain.,IBSAL (Instituto de investigación Biomédica de Salamanca), Salamanca, Spain
| | - Carla Ijurko
- Department of Biochemistry and Molecular Biology, University of Salamanca, Lab. 122, Edificio Departamental, Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain.,IBSAL (Instituto de investigación Biomédica de Salamanca), Salamanca, Spain
| | - Ángel Hernández-Hernández
- Department of Biochemistry and Molecular Biology, University of Salamanca, Lab. 122, Edificio Departamental, Plaza Doctores de la Reina s/n, 37007, Salamanca, Spain. .,IBSAL (Instituto de investigación Biomédica de Salamanca), Salamanca, Spain.
| |
Collapse
|
140
|
Huang K, Ma K, Hung Y, Lo L, Lin K, Liu P, Hu M, Chueh S. A new copper ionophore DPMQ protects cells against ultraviolet B irradiation by inhibiting the TRPV1 channel. J Cell Physiol 2018; 233:9594-9610. [DOI: 10.1002/jcp.26861] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/23/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Kuo‐Feng Huang
- Division of Plastic Surgery, Department of Surgery Chi Mei Medical Center Tainan Taiwan Republic of China
| | - Kuo‐Hsing Ma
- Department of Biology and Anatomy National Defense Medical Center Taipei Taiwan Republic of China
| | - Yu‐Chien Hung
- Department of Biochemistry National Defense Medical Center Taipei Taiwan Republic of China
| | - Liang‐Chuan Lo
- Department of Biochemistry National Defense Medical Center Taipei Taiwan Republic of China
| | - Kuo‐Chen Lin
- Department of Biochemistry National Defense Medical Center Taipei Taiwan Republic of China
| | - Pei‐Shan Liu
- Department of Microbiology Soochow University Taipei Taiwan Republic of China
| | - Ming‐Kuan Hu
- Department of Medicinal Chemistry School of Pharmacy, National Defense Medical Center Taipei Taiwan Republic of China
| | - Sheau‐Huei Chueh
- Department of Biochemistry National Defense Medical Center Taipei Taiwan Republic of China
| |
Collapse
|
141
|
Vultur A, Gibhardt CS, Stanisz H, Bogeski I. The role of the mitochondrial calcium uniporter (MCU) complex in cancer. Pflugers Arch 2018; 470:1149-1163. [PMID: 29926229 DOI: 10.1007/s00424-018-2162-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/14/2018] [Accepted: 05/30/2018] [Indexed: 01/07/2023]
Abstract
The important role of mitochondria in cancer biology is gaining momentum. With their regulation of cell survival, metabolism, basic cell building blocks, and immunity, among other functions, mitochondria affect not only cancer progression but also the response and resistance to current treatments. Calcium ions are constantly shuttled in and out of mitochondria; thus, playing an important role in the regulation of various cellular processes. The mitochondrial calcium uniporter (MCU) channel and its associated regulators transport calcium across the inner mitochondrial membrane to the mitochondrial matrix. Due to this central role and the capacity to affect cell behavior and fate, the MCU complex is being investigated in different cancers and cancer-related conditions. Here, we review current knowledge on the role of the MCU complex in multiple cancer types and models; we also provide a perspective for future research and clinical considerations.
Collapse
Affiliation(s)
- Adina Vultur
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Georg-August-University, Humboldtallee 23, 37073, Göttingen, Germany
| | - Christine S Gibhardt
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Georg-August-University, Humboldtallee 23, 37073, Göttingen, Germany
| | - Hedwig Stanisz
- Department of Dermatology, Venereology and Allergology, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Ivan Bogeski
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Georg-August-University, Humboldtallee 23, 37073, Göttingen, Germany.
| |
Collapse
|
142
|
Paupe V, Prudent J. New insights into the role of mitochondrial calcium homeostasis in cell migration. Biochem Biophys Res Commun 2018; 500:75-86. [PMID: 28495532 PMCID: PMC5930976 DOI: 10.1016/j.bbrc.2017.05.039] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 05/07/2017] [Indexed: 01/23/2023]
Abstract
Mitochondria are dynamic organelles involved in numerous physiological functions. Beyond their function in ATP production, mitochondria regulate cell death, reactive oxygen species (ROS) generation, immunity and metabolism. Mitochondria also play a key role in the buffering of cytosolic calcium, and calcium transported into the matrix regulates mitochondrial metabolism. Recently, the identification of the mitochondrial calcium uniporter (MCU) and associated regulators has allowed the characterization of new physiological roles for calcium in both mitochondrial and cellular homeostasis. Indeed, recent work has highlighted the importance of mitochondrial calcium homeostasis in regulating cell migration. Cell migration is a property common to all metazoans and is critical to embryogenesis, cancer progression, wound-healing and immune surveillance. Previous work has established that cytoplasmic calcium is a key regulator of cell migration, as oscillations in cytosolic calcium activate cytoskeletal remodelling, actin contraction and focal adhesion (FA) turnover necessary for cell movement. Recent work using animal models and in cellulo experiments to genetically modulate MCU and partners have shed new light on the role of mitochondrial calcium dynamics in cytoskeletal remodelling through the modulation of ATP and ROS production, as well as intracellular calcium signalling. This review focuses on MCU and its regulators in cell migration during physiological and pathophysiological processes including development and cancer. We also present hypotheses to explain the molecular mechanisms by which MCU may regulate mitochondrial dynamics and motility to drive cell migration.
Collapse
Affiliation(s)
- Vincent Paupe
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, United Kingdom
| | - Julien Prudent
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, United Kingdom.
| |
Collapse
|
143
|
Mammucari C, Raffaello A, Vecellio Reane D, Gherardi G, De Mario A, Rizzuto R. Mitochondrial calcium uptake in organ physiology: from molecular mechanism to animal models. Pflugers Arch 2018. [PMID: 29541860 PMCID: PMC6060757 DOI: 10.1007/s00424-018-2123-2] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mitochondrial Ca2+ is involved in heterogeneous functions, ranging from the control of metabolism and ATP production to the regulation of cell death. In addition, mitochondrial Ca2+ uptake contributes to cytosolic [Ca2+] shaping thus impinging on specific Ca2+-dependent events. Mitochondrial Ca2+ concentration is controlled by influx and efflux pathways: the former controlled by the activity of the mitochondrial Ca2+ uniporter (MCU), the latter by the Na+/Ca2+ exchanger (NCLX) and the H+/Ca2+ (mHCX) exchanger. The molecular identities of MCU and of NCLX have been recently unraveled, thus allowing genetic studies on their physiopathological relevance. After a general framework on the significance of mitochondrial Ca2+ uptake, this review discusses the structure of the MCU complex and the regulation of its activity, the importance of mitochondrial Ca2+ signaling in different physiological settings, and the consequences of MCU modulation on organ physiology.
Collapse
Affiliation(s)
| | - Anna Raffaello
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | | | - Gaia Gherardi
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Agnese De Mario
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padova, Padua, Italy.
| |
Collapse
|
144
|
A novel tetrahydroisoquinoline (THIQ) analogue induces mitochondria-dependent apoptosis. Eur J Med Chem 2018; 150:719-728. [PMID: 29573707 DOI: 10.1016/j.ejmech.2018.03.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/04/2018] [Accepted: 03/05/2018] [Indexed: 12/31/2022]
Abstract
Lung cancer continues to be a leading cause of cancer-related death worldwide, with non-small cell lung cancer (NSCLC) accounting for more than 80% of lung cancer cases. Current therapies for NSCLC have only limited effect and treatment resistance develops rapidly. In a previous study, we have shown that C1-phenylethynyl tetrahydroisoquinoline (THIQ) analogue 4 has anti-proliferative activity against PC3 human prostate cancer cells. However, this anticancer effect was achieved with relatively high IC50 in A549 lung cancer cells. To improve the potency of the drug, in the present study, a series of novel THIQ analogues (analogues 5a-d) were prepared by using an oxidative C-H functionalization strategy, and their potential anticancer activities on A549 lung cancer cells were investigated. Among these analogues, analogue 5c can markedly inhibit A549 cell proliferation in a dose-dependent manner with a reasonable IC50 of 14.61 ± 1.03 μM. This effect was mediated by analogue 5c-induced G0/G1 phase arrest and cell apoptosis. Treatment with analogue 5c was shown to induce reactive oxygen species (ROS) accumulation, disruption of mitochondrial membrane potential, reduction of glutathione, elevation of intracellular calcium ion (Ca2+), and activation of Caspase-3. Furthermore, analogue 5c can lead to DNA double-strand break and the activation of p53 pathway in A549 cells. In conclusion, the oxidative C-H functionalization strategy to generate analogue 5c could improve the drug anticancer efficacy by inducing mitochondria-dependent apoptosis in A549 cells.
Collapse
|
145
|
Li X, Wang B, Tang L, Zhang Y, Chen L, Gu L, Zhang F, Ouyang J, Zhang X. GSTA1 Expression Is Correlated With Aldosterone Level in KCNJ5-Mutated Adrenal Aldosterone-Producing Adenoma. J Clin Endocrinol Metab 2018; 103:813-823. [PMID: 29165597 DOI: 10.1210/jc.2017-00950] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 11/15/2017] [Indexed: 01/08/2023]
Abstract
CONTEXT KCNJ5 mutation is a major cause of aldosterone-producing adenomas (APAs). The development of APA apart from KCNJ5 mutation is less investigated. OBJECTIVE To investigate other mechanisms affecting aldosterone secretion apart from KCNJ5. PATIENTS AND METHODS Six pairs of KCNJ5-mutated, high and low aldosterone-secreting APAs, five non-KCNJ5-mutated APAs, and four normal adrenal glands were assayed by Affymetrix GeneChip Human Transcriptome Array 2.0. A total of 113 APA samples were investigated to explore the expression of glutathione-S-transferase A1 (GSTA1). H295R cells were used to verify the function of GSTA1. RESULTS GSTA1 was the top gene downregulated in high-aldosterone KCNJ5-mutated APAs. GSTA1 was also downregulated in KCNJ5-mutated APAs compared with wild-type KCNJ5 APAs. Accordingly, mutant KCNJ5 decreased GSTA1 messenger RNA and protein expression levels. GSTA1 overexpression suppressed aldosterone secretion whether in wild-type or mutant KCNJ5 H295R cells. Adding ethacrynic acid or silencing of GSTA1 increased aldosterone secretion by increasing reactive oxygen species (ROS), superoxide, H2O2 levels, and Ca2+ influx. The expression of the transcription factors NR4A1, NR4A2, and CAMK1 and intracellular Ca2+ were significantly upregulated by GSTA1 inhibition. The reduced form of NAD phosphate oxidase inhibitor or H2O2 scavenger or blocking calmodulin or calcium channels could significantly reduce aldosterone secretion in GSTA1-inhibited cells. CONCLUSIONS (1) GSTA1 expression is reversely correlated with aldosterone level in KCNJ5-mutated APAs, (2) GSTA1 regulates aldosterone secretion by ROS and Ca2+ signaling, and (3) KCNJ5 mutation downregulates GSTA1 expression, and overexpression of GSTA1 reverses increased aldosterone in KCNJ5-mutated adrenal cells.
Collapse
Affiliation(s)
- Xintao Li
- State Key Laboratory of Kidney Disease, Department of Urology, Chinese People's Liberation Army Medical Academy, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Baojun Wang
- State Key Laboratory of Kidney Disease, Department of Urology, Chinese People's Liberation Army Medical Academy, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Lu Tang
- State Key Laboratory of Kidney Disease, Department of Urology, Chinese People's Liberation Army Medical Academy, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yu Zhang
- State Key Laboratory of Kidney Disease, Department of Urology, Chinese People's Liberation Army Medical Academy, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Luyao Chen
- State Key Laboratory of Kidney Disease, Department of Urology, Chinese People's Liberation Army Medical Academy, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Liangyou Gu
- State Key Laboratory of Kidney Disease, Department of Urology, Chinese People's Liberation Army Medical Academy, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Fan Zhang
- State Key Laboratory of Kidney Disease, Department of Urology, Chinese People's Liberation Army Medical Academy, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jinzhi Ouyang
- Department of Outpatient Officer Consultation Room, Chinese People's Liberation Army Medical Academy, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xu Zhang
- State Key Laboratory of Kidney Disease, Department of Urology, Chinese People's Liberation Army Medical Academy, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
146
|
Proscillaridin A Promotes Oxidative Stress and ER Stress, Inhibits STAT3 Activation, and Induces Apoptosis in A549 Lung Adenocarcinoma Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3853409. [PMID: 29576846 PMCID: PMC5821950 DOI: 10.1155/2018/3853409] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/21/2017] [Accepted: 11/16/2017] [Indexed: 12/13/2022]
Abstract
Cardiac glycosides are natural compounds used for the treatment of cardiovascular disorders. Although originally prescribed for cardiovascular diseases, more recently, they have been rediscovered for their potential use in the treatment of cancer. Proscillaridin A (PSD-A), a cardiac glycoside component of Urginea maritima, has been reported to exhibit anticancer activity. However, the cellular targets and anticancer mechanism of PSD-A in various cancers including lung cancer remain largely unexplored. In the present study, we found that PSD-A inhibits growth and induces apoptosis in A549 lung adenocarcinoma cells. The anticancer activity of PSD-A was found to be associated with the activation of JNK, induction of ER stress, mitochondrial dysfunction, and inhibition of STAT3 activation. PSD-A induces oxidative stress as evidenced from ROS generation, GSH depletion, and decreased activity of TrxR1. PSD-A-mediated ER stress was verified by increased phosphorylation of eIF2α and expression of its downstream effector proteins ATF4, CHOP, and caspases-4. PSD-A triggered apoptosis by inducing JNK (1/2) activation, increasing bax/bcl-2 ratio, dissipating mitochondrial membrane potential, and inducing cleavage of caspases and PARP. Further study revealed that PSD-A inhibits both constitutive and inducible STAT3 activations and decreases STAT3 DNA-binding activity. Moreover, PSD-A-mediated inhibition of STAT3 activation was found to be associated with increased SHP-1 expression, decreased phosphorylation of Src, and binding of PSD-A with STAT3 SH2 domain. Finally, STAT3 knockdown by shRNA inhibited growth and enhanced apoptotic efficacy of PSD-A. Taken together, the data suggest that PSD-A could be developed into a potential therapeutic agent against lung adenocarcinoma.
Collapse
|
147
|
Krylatov AV, Maslov LN, Voronkov NS, Boshchenko AA, Popov SV, Gomez L, Wang H, Jaggi AS, Downey JM. Reactive Oxygen Species as Intracellular Signaling Molecules in the Cardiovascular System. Curr Cardiol Rev 2018; 14:290-300. [PMID: 29962348 PMCID: PMC6300799 DOI: 10.2174/1573403x14666180702152436] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 06/13/2018] [Accepted: 06/20/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Redox signaling plays an important role in the lives of cells. This signaling not only becomes apparent in pathologies but is also thought to be involved in maintaining physiological homeostasis. Reactive Oxygen Species (ROS) can activate protein kinases: CaMKII, PKG, PKA, ERK, PI3K, Akt, PKC, PDK, JNK, p38. It is unclear whether it is a direct interaction of ROS with these kinases or whether their activation is a consequence of inhibition of phosphatases. ROS have a biphasic effect on the transport of Ca2+ in the cell: on one hand, they activate the sarcoplasmic reticulum Ca2+-ATPase, which can reduce the level of Ca2+ in the cell, and on the other hand, they can inactivate Ca2+-ATPase of the plasma membrane and open the cation channels TRPM2, which promote Ca2+-loading and subsequent apoptosis. ROS inhibit the enzyme PHD2, which leads to the stabilization of HIF-α and the formation of the active transcription factor HIF. CONCLUSION Activation of STAT3 and STAT5, induced by cytokines or growth factors, may include activation of NADPH oxidase and enhancement of ROS production. Normal physiological production of ROS under the action of cytokines activates the JAK/STAT while excessive ROS production leads to their inhibition. ROS cause the activation of the transcription factor NF-κB. Physiological levels of ROS control cell proliferation and angiogenesis. ROS signaling is also involved in beneficial adaptations to survive ischemia and hypoxia, while further increases in ROS can trigger programmed cell death by the mechanism of apoptosis or autophagy. ROS formation in the myocardium can be reduced by moderate exercise.
Collapse
Affiliation(s)
| | - Leonid N. Maslov
- Address correspondence to this author at the Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of
Science, Tomsk, Russia; Tel: 3822 262174; Fax: 3822 555057;
E-mail:
| | | | | | | | | | | | | | | |
Collapse
|
148
|
Redox-Dependent Circuits Regulating B Lymphocyte Physiology. Immunology 2018. [DOI: 10.1016/b978-0-12-809819-6.00013-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
149
|
Alevriadou BR, Shanmughapriya S, Patel A, Stathopulos PB, Madesh M. Mitochondrial Ca 2+ transport in the endothelium: regulation by ions, redox signalling and mechanical forces. J R Soc Interface 2017; 14:rsif.2017.0672. [PMID: 29237825 DOI: 10.1098/rsif.2017.0672] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 11/16/2017] [Indexed: 02/07/2023] Open
Abstract
Calcium (Ca2+) transport by mitochondria is an important component of the cell Ca2+ homeostasis machinery in metazoans. Ca2+ uptake by mitochondria is a major determinant of bioenergetics and cell fate. Mitochondrial Ca2+ uptake occurs via the mitochondrial Ca2+ uniporter (MCU) complex, an inner mitochondrial membrane protein assembly consisting of the MCU Ca2+ channel, as its core component, and the MCU complex regulatory/auxiliary proteins. In this review, we summarize the current knowledge on the molecular nature of the MCU complex and its regulation by intra- and extramitochondrial levels of divalent ions and reactive oxygen species (ROS). Intracellular Ca2+ concentration ([Ca2+]i), mitochondrial Ca2+ concentration ([Ca2+]m) and mitochondrial ROS (mROS) are intricately coupled in regulating MCU activity. Here, we highlight the contribution of MCU activity to vascular endothelial cell (EC) function. Besides the ionic and oxidant regulation, ECs are continuously exposed to haemodynamic forces (either pulsatile or oscillatory fluid mechanical shear stresses, depending on the precise EC location within the arteries). Thus, we also propose an EC mechanotransduction-mediated regulation of MCU activity in the context of vascular physiology and atherosclerotic vascular disease.
Collapse
Affiliation(s)
- B Rita Alevriadou
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA .,Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH 43210, USA.,Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Santhanam Shanmughapriya
- Department of Medical Genetics and Molecular Biochemistry, Temple University, Philadelphia, PA 19140, USA.,Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Akshar Patel
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA.,Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH 43210, USA.,Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Peter B Stathopulos
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada N6A 5C1
| | - Muniswamy Madesh
- Department of Medical Genetics and Molecular Biochemistry, Temple University, Philadelphia, PA 19140, USA .,Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
150
|
Takata N, Ohshima Y, Suzuki-Karasaki M, Yoshida Y, Tokuhashi Y, Suzuki-Karasaki Y. Mitochondrial Ca2+ removal amplifies TRAIL cytotoxicity toward apoptosis-resistant tumor cells via promotion of multiple cell death modalities. Int J Oncol 2017; 51:193-203. [PMID: 28560396 DOI: 10.3892/ijo.2017.4020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 05/16/2017] [Indexed: 11/06/2022] Open
Abstract
Ca2+ has emerged as a new target for cancer treatment since tumor-specific traits in Ca2+ dynamics contributes to tumorigenesis, malignant phenotypes, drug resistance, and survival in different tumor types. However, Ca2+ has a dual (pro-death and pro-survival) function in tumor cells depending on the experimental conditions. Therefore, it is necessary to minimize the onset of the pro-survival Ca2+ signals caused by the therapy. For this purpose, a better understanding of pro-survival Ca2+ pathways in cancer cells is critical. Here we report that Ca2+ protects malignant melanoma (MM) and osteosarcoma (OS) cells from tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) cytotoxicity. Simultaneous measurements using the site-specific Ca2+ probes showed that acute TRAIL treatment rapidly and dose-dependently increased the cytosolic Ca2+ concentration ([Ca2+]cyt) and mitochondrial Ca2+ concentration ([Ca2+]mit) Pharmacological analyses revealed that the [Ca2+]mit remodeling was under control of mitochondrial Ca2+ uniporter (MCU), mitochondrial permeability transition pore (MPTP), and a Ca2+ transport pathway sensitive to capsazepine and AMG9810. Ca2+ chelators and the MCU inhibitor ruthenium 360, an MPTP opener atractyloside, capsazepine, and AMG9810 all decreased [Ca2+]mit and sensitized these tumor cells to TRAIL cytotoxicity. The Ca2+ modulation enhanced both apoptotic and non-apoptotic cell death. Although the [Ca2+]mit reduction potentiated TRAIL-induced caspase-3/7 activation and cell membrane damage within 24 h, this potentiation of cell death became pronounced at 72 h, and not blocked by caspase inhibition. Our findings suggest that in MM and OS cells mitochondrial Ca2+ removal can promote apoptosis and non-apoptotic cell death induction by TRAIL. Therefore, mitochondrial Ca2+ removal can be exploited to overcome the resistance of these cancers to TRAIL.
Collapse
Affiliation(s)
- Natsuhiko Takata
- Department of Orthopedic Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Yohei Ohshima
- Department of Orthopedic Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Miki Suzuki-Karasaki
- Department of Orthopedic Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Yukihiro Yoshida
- Department of Orthopedic Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Yasuaki Tokuhashi
- Department of Orthopedic Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | | |
Collapse
|