1
|
Song G, Yang Z, Cheng J, Lin H, Huang Y, Lv F, Bai H, Wang S. Chemiluminescence Resonance Energy Transfer for Targeted Photoactivation of Ion Channels in Living Cells. Angew Chem Int Ed Engl 2025:e202423792. [PMID: 39888216 DOI: 10.1002/anie.202423792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/01/2025]
Abstract
The regulation of oxidative stress in living cells is essential for maintaining cellular processes and signal transduction. However, developing straightforward strategies to activate oxidative stress-sensitive membrane channels in situ poses significant challenges. In this study, we present a chemiluminescence resonance energy transfer (CRET) system based on a conjugated oligomer, oligo(p-phenylenevinylene)-imidazolium (OPV-Im), designed for the activation of transient receptor potential melastatin 2 (TRPM2) calcium channels in situ by superoxide anion (O2⋅-) without requiring external light sources. The OPV-Im oligomer targeted the cell membrane efficiently, leading to the activation of TRPM2 channels in situ by the CRET process and subsequent intracellular calcium overload. This cascade resulted in mitochondrial damage and inhibition of autophagy, ultimately inducing cell apoptosis. Additionally, this strategy could be applied for the selective killing of tumor cells that overexpress TRPM2 ion channels and for inhibiting the growth of three-dimensional (3D) tumor spheroids. Our system offers a novel approach for regulating ion channel activity and oxidative stress in living cells compared to optogenetics and photodynamic therapy.
Collapse
Affiliation(s)
- Gang Song
- Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences., Beijing, 100049, P. R. China
| | - Zhiwen Yang
- Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences., Beijing, 100049, P. R. China
| | - Junjie Cheng
- Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences., Beijing, 100049, P. R. China
| | - Hongrui Lin
- Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences., Beijing, 100049, P. R. China
| | - Yiming Huang
- Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Fengting Lv
- Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Haotian Bai
- Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Shu Wang
- Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences., Beijing, 100049, P. R. China
| |
Collapse
|
2
|
Mandlem VKK, Rivera A, Khan Z, Quazi SH, Deba F. TLR4 induced TRPM2 mediated neuropathic pain. Front Pharmacol 2024; 15:1472771. [PMID: 39329114 PMCID: PMC11424904 DOI: 10.3389/fphar.2024.1472771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/02/2024] [Indexed: 09/28/2024] Open
Abstract
Ion channels play an important role in mediating pain through signal transduction, regulation, and control of responses, particularly in neuropathic pain. Transient receptor potential channel superfamily plays an important role in cation permeability and cellular signaling. Transient receptor potential channel Melastatin 2 (TRPM2) subfamily regulates Ca2+ concentration in response to various chemicals and signals from the surrounding environment. TRPM2 has a role in several physiological functions such as cellular osmosis, temperature sensing, cellular proliferation, as well as the manifestation of many disease processes such as pain process, cancer, apoptosis, endothelial dysfunction, angiogenesis, renal and lung fibrosis, and cerebral ischemic stroke. Toll-like Receptor 4 (TLR4) is a critical initiator of the immune response to inflammatory stimuli, particularly those triggered by Lipopolysaccharide (LPS). It activates downstream pathways leading to the production of oxidative molecules and inflammatory cytokines, which are modulated by basal and store-operated calcium ion signaling. The cytokine production and release cause an imbalance of antioxidant enzymes and redox potential in the Endoplasmic Reticulum and mitochondria due to oxidative stress, which results from TLR-4 activation and consequently induces the production of inflammatory cytokines in neuronal cells, exacerbating the pain process. Very few studies have reported the role of TRPM2 and its association with Toll-like receptors in the context of neuropathic pain. However, the molecular mechanism underlying the interaction between TRPM2 and TLR-4 and the quantum of impact in acute and chronic neuropathic pain remains unclear. Understanding the link between TLR-4 and TRPM2 will provide more insights into pain regulation mechanisms for the development of new therapeutic molecules to address neuropathic pain.
Collapse
Affiliation(s)
- Venkata Kiran Kumar Mandlem
- Departmental of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX, United States
| | - Ana Rivera
- Departmental of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX, United States
| | - Zaina Khan
- Departmental of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX, United States
- Departmental of Neuroscience, University of Texas at Dallas, Richardson, TX, United States
| | - Sohel H Quazi
- Departmental of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX, United States
- Department of Biology, Division of Natural and Computation Sciences, Texas College, Tyler, TX, United States
| | - Farah Deba
- Departmental of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX, United States
| |
Collapse
|
3
|
Benzi A, Heine M, Spinelli S, Salis A, Worthmann A, Diercks B, Astigiano C, Pérez Mato R, Memushaj A, Sturla L, Vellone V, Damonte G, Jaeckstein MY, Koch-Nolte F, Mittrücker HW, Guse AH, De Flora A, Heeren J, Bruzzone S. The TRPM2 ion channel regulates metabolic and thermogenic adaptations in adipose tissue of cold-exposed mice. Front Endocrinol (Lausanne) 2024; 14:1251351. [PMID: 38390373 PMCID: PMC10882718 DOI: 10.3389/fendo.2023.1251351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 11/16/2023] [Indexed: 02/24/2024] Open
Abstract
Introduction During thermogenesis, adipose tissue (AT) becomes more active and enhances oxidative metabolism. The promotion of this process in white AT (WAT) is called "browning" and, together with the brown AT (BAT) activation, is considered as a promising approach to counteract obesity and metabolic diseases. Transient receptor potential cation channel, subfamily M, member 2 (TRPM2), is an ion channel that allows extracellular Ca2+ influx into the cytosol, and is gated by adenosine diphosphate ribose (ADPR), produced from NAD+ degradation. The aim of this study was to investigate the relevance of TRPM2 in the regulation of energy metabolism in BAT, WAT, and liver during thermogenesis. Methods Wild type (WT) and Trpm2-/- mice were exposed to 6°C and BAT, WAT and liver were collected to evaluate mRNA, protein levels and ADPR content. Furthermore, O2 consumption, CO2 production and energy expenditure were measured in these mice upon thermogenic stimulation. Finally, the effect of the pharmacological inhibition of TRPM2 was assessed in primary adipocytes, evaluating the response upon stimulation with the β-adrenergic receptor agonist CL316,243. Results Trpm2-/- mice displayed lower expression of browning markers in AT and lower energy expenditure in response to thermogenic stimulus, compared to WT animals. Trpm2 gene overexpression was observed in WAT, BAT and liver upon cold exposure. In addition, ADPR levels and mono/poly-ADPR hydrolases expression were higher in mice exposed to cold, compared to control mice, likely mediating ADPR generation. Discussion Our data indicate TRPM2 as a fundamental player in BAT activation and WAT browning. TRPM2 agonists may represent new pharmacological strategies to fight obesity.
Collapse
Affiliation(s)
- Andrea Benzi
- Department of Experimental Medicine-Section of Biochemistry, University of Genova, Genova, Italy
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sonia Spinelli
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Annalisa Salis
- Department of Experimental Medicine-Section of Biochemistry, University of Genova, Genova, Italy
| | - Anna Worthmann
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Björn Diercks
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cecilia Astigiano
- Department of Experimental Medicine-Section of Biochemistry, University of Genova, Genova, Italy
| | - Raúl Pérez Mato
- Department of Experimental Medicine-Section of Biochemistry, University of Genova, Genova, Italy
| | - Adela Memushaj
- Department of Experimental Medicine-Section of Biochemistry, University of Genova, Genova, Italy
| | - Laura Sturla
- Department of Experimental Medicine-Section of Biochemistry, University of Genova, Genova, Italy
| | - Valerio Vellone
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genova, Italy
- Pathology Unit, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Gianluca Damonte
- Department of Experimental Medicine-Section of Biochemistry, University of Genova, Genova, Italy
| | - Michelle Y Jaeckstein
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Willi Mittrücker
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas H Guse
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Antonio De Flora
- Department of Experimental Medicine-Section of Biochemistry, University of Genova, Genova, Italy
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Santina Bruzzone
- Department of Experimental Medicine-Section of Biochemistry, University of Genova, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
4
|
Okada Y, Numata T, Sabirov RZ, Kashio M, Merzlyak PG, Sato-Numata K. Cell death induction and protection by activation of ubiquitously expressed anion/cation channels. Part 3: the roles and properties of TRPM2 and TRPM7. Front Cell Dev Biol 2023; 11:1246955. [PMID: 37842082 PMCID: PMC10576435 DOI: 10.3389/fcell.2023.1246955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
Cell volume regulation (CVR) is a prerequisite for animal cells to survive and fulfill their functions. CVR dysfunction is essentially involved in the induction of cell death. In fact, sustained normotonic cell swelling and shrinkage are associated with necrosis and apoptosis, and thus called the necrotic volume increase (NVI) and the apoptotic volume decrease (AVD), respectively. Since a number of ubiquitously expressed ion channels are involved in the CVR processes, these volume-regulatory ion channels are also implicated in the NVI and AVD events. In Part 1 and Part 2 of this series of review articles, we described the roles of swelling-activated anion channels called VSOR or VRAC and acid-activated anion channels called ASOR or PAC in CVR and cell death processes. Here, Part 3 focuses on therein roles of Ca2+-permeable non-selective TRPM2 and TRPM7 cation channels activated by stress. First, we summarize their phenotypic properties and molecular structure. Second, we describe their roles in CVR. Since cell death induction is tightly coupled to dysfunction of CVR, third, we focus on their participation in the induction of or protection against cell death under oxidative, acidotoxic, excitotoxic, and ischemic conditions. In this regard, we pay attention to the sensitivity of TRPM2 and TRPM7 to a variety of stress as well as to their capability to physicall and functionally interact with other volume-related channels and membrane enzymes. Also, we summarize a large number of reports hitherto published in which TRPM2 and TRPM7 channels are shown to be involved in cell death associated with a variety of diseases or disorders, in some cases as double-edged swords. Lastly, we attempt to describe how TRPM2 and TRPM7 are organized in the ionic mechanisms leading to cell death induction and protection.
Collapse
Affiliation(s)
- Yasunobu Okada
- National Institute for Physiological Sciences (NIPS), Okazaki, Japan
- Department of Integrative Physiology, Graduate School of Medicine, AkitaUniversity, Akita, Japan
- Department of Physiology, School of Medicine, Aichi Medical Uniersity, Nagakute, Japan
- Department of Physiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Cardiovascular Research Institute, Yokohama City University, Yokohama, Japan
| | - Tomohiro Numata
- Department of Integrative Physiology, Graduate School of Medicine, AkitaUniversity, Akita, Japan
| | - Ravshan Z. Sabirov
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Makiko Kashio
- National Institute for Physiological Sciences (NIPS), Okazaki, Japan
- Department of Physiology, School of Medicine, Aichi Medical Uniersity, Nagakute, Japan
| | - Peter G. Merzlyak
- Institute of Biophysics and Biochemistry, National University of Uzbekistan, Tashkent, Uzbekistan
| | - Kaori Sato-Numata
- Department of Integrative Physiology, Graduate School of Medicine, AkitaUniversity, Akita, Japan
| |
Collapse
|
5
|
Ali ES, Chakrabarty B, Ramproshad S, Mondal B, Kundu N, Sarkar C, Sharifi-Rad J, Calina D, Cho WC. TRPM2-mediated Ca 2+ signaling as a potential therapeutic target in cancer treatment: an updated review of its role in survival and proliferation of cancer cells. Cell Commun Signal 2023; 21:145. [PMID: 37337283 DOI: 10.1186/s12964-023-01149-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 04/28/2023] [Indexed: 06/21/2023] Open
Abstract
The transient receptor potential melastatin subfamily member 2 (TRPM2), a thermo and reactive oxygen species (ROS) sensitive Ca2+-permeable cation channel has a vital role in surviving the cell as well as defending the adaptability of various cell groups during and after oxidative stress. It shows higher expression in several cancers involving breast, pancreatic, prostate, melanoma, leukemia, and neuroblastoma, indicating it raises the survivability of cancerous cells. In various cancers including gastric cancers, and neuroblastoma, TRPM2 is known to conserve viability, and several underlying mechanisms of action have been proposed. Transcription factors are thought to activate TRPM2 channels, which is essential for cell proliferation and survival. In normal physiological conditions with an optimal expression of TRPM2, mitochondrial ROS is produced in optimal amounts while regulation of antioxidant expression is carried on. Depletion of TRPM2 overexpression or activity has been shown to improve ischemia-reperfusion injury in organ levels, reduce tumor growth and/or viability of various malignant cancers like breast, gastric, pancreatic, prostate, head and neck cancers, melanoma, neuroblastoma, T-cell and acute myelogenous leukemia. This updated and comprehensive review also analyzes the mechanisms by which TRPM2-mediated Ca2+ signaling can regulate the growth and survival of different types of cancer cells. Based on the discussion of the available data, it can be concluded that TRPM2 may be a unique therapeutic target in the treatment of several types of cancer. Video Abstract.
Collapse
Affiliation(s)
- Eunus S Ali
- College of Medicine and Public Health, Flinders University, Bedford Park, 5042, Australia
- Gaco Pharmaceuticals, Dhaka, 1000, Bangladesh
- Present Address: Department of Biochemistry and Molecular Genetics, and Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, 303 E Superior St, Chicago, IL, 60611, USA
| | | | - Sarker Ramproshad
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj, 1400, Bangladesh
| | - Banani Mondal
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj, 1400, Bangladesh
| | - Neloy Kundu
- Pharmacy Discipline, Khulna University, Khulna, 9208, Bangladesh
| | - Chandan Sarkar
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | | | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, 200349, Romania.
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China.
| |
Collapse
|
6
|
Yang H, Tai F, Wang T, Zheng X, Ge C, Qin Y, Fu H. Hydrogen peroxide and iron ions can modulate lipid peroxidation, apoptosis, and the cell cycle, but do not have a significant effect on DNA double-strand break. Biochem Biophys Res Commun 2023; 651:121-126. [PMID: 36822125 DOI: 10.1016/j.bbrc.2023.02.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/17/2023] [Accepted: 02/09/2023] [Indexed: 02/11/2023]
Abstract
Hydroxyl radical (·OH) generated by the Fenton reaction between transition metal ions and hydrogen peroxide (H2O2) can induce significant cellular damage. However, the specific mechanism of ·OH-induced cell death has not been systematically studied. In this study, we reacted FeSO4 and Fe3O4 magnetic nanoparticles with H2O2 and found that ·OH generated from the intracellular Fenton reaction can lead to significant cell death. The Fenton reaction between Fe2+ with H2O2 resulted in a shift in lipid peroxidation and cell cycle arrest. It is noteworthy that the ·OH generated from the Fenton reaction triggered severe apoptosis but did not lead to DNA double-strand breakage. Our results suggest that the Fenton reaction had acute cytotoxicity, which was primarily due to ·OH produced from the Fenton reaction inducing lipid peroxidation and apoptosis and modulating the cell cycle, but not by inducing DNA damage.
Collapse
Affiliation(s)
- Hexi Yang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, China; Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Fumin Tai
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Tiantian Wang
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Xiaofei Zheng
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Changhui Ge
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Yide Qin
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, China.
| | - Hanjiang Fu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230032, China; Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| |
Collapse
|
7
|
Distribution and Assembly of TRP Ion Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1349:111-138. [PMID: 35138613 DOI: 10.1007/978-981-16-4254-8_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the last several decades, a large family of ion channels have been identified and studied intensively as cellular sensors for diverse physical and/or chemical stimuli. Named transient receptor potential (TRP) channels, they play critical roles in various aspects of cellular physiology. A large number of human hereditary diseases are found to be linked to TRP channel mutations, and their dysregulations lead to acute or chronical health problems. As TRP channels are named and categorized mostly based on sequence homology rather than functional similarities, they exhibit substantial functional diversity. Rapid advances in TRP channel study have been made in recent years and reported in a vast body of literature; a summary of the latest advancements becomes necessary. This chapter offers an overview of current understandings of TRP channel distribution and subunit assembly.
Collapse
|
8
|
Jammoul M, Lawand N. Melatonin: a Potential Shield against Electromagnetic Waves. Curr Neuropharmacol 2021; 20:648-660. [PMID: 34635042 DOI: 10.2174/1570159x19666210609163946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/16/2021] [Accepted: 05/16/2021] [Indexed: 11/22/2022] Open
Abstract
Melatonin, a vital hormone synthesized by the pineal gland, has been implicated in various physiological functions and in circadian rhythm regulation. Its role in the protection against the non-ionizing electromagnetic field (EMF), known to disrupt the body's oxidative/anti-oxidative balance, has been called into question due to inconsistent results observed across studies. This review provides the current state of knowledge on the interwoven relationship between melatonin, EMF, and oxidative stress. Based on synthesized evidence, we present a model that best describes the mechanisms underlying the protective effects of melatonin against RF/ELF-EMF induced oxidative stress. We show that the free radical scavenger activity of melatonin is enabled through reduction of the radical pair singlet-triplet conversion rate and the concentration of the triplet products. Moreover, this review aims to highlight the potential therapeutic benefits of melatonin against the detrimental effects of EMF, in general, and electromagnetic hypersensitivity (EHS), in particular.
Collapse
Affiliation(s)
- Maya Jammoul
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut. Lebanon
| | - Nada Lawand
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut. Lebanon
| |
Collapse
|
9
|
Hung CY, Tan CH. TRP Channels in Nociception and Pathological Pain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1099:13-27. [PMID: 30306511 DOI: 10.1007/978-981-13-1756-9_2] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Thermal and noxious stimuli are detected by specialized nerve endings, which transform the stimuli into electrical signals and transmit the signals into central nervous system to facilitate the perception of temperature and pain. Several members within the transient receptor potential (TRP) channel family serve as the sensors for temperature and noxious stimuli and are involved in the development of pathological pain, especially inflammatory pain. Various inflammatory mediators can sensitize and modulate the activation threshold of TRP channels and result in the development of inflammatory pain behaviors. A brief review of the role of TRP channels in nociception and the modulatory mechanisms of TRP channels by inflammatory mediators, focusing on TRPV1, TRPA1, and TRPM2, will be presented. Recent advances in the development of therapeutic strategies targeting against TRP channels will also be reviewed.
Collapse
Affiliation(s)
- Chen-Yu Hung
- Department of General Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chun-Hsiang Tan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
10
|
Eraslan E, Tanyeli A, Polat E, Polat E. 8-Br-cADPR, a TRPM2 ion channel antagonist, inhibits renal ischemia-reperfusion injury. J Cell Physiol 2018; 234:4572-4581. [PMID: 30191993 DOI: 10.1002/jcp.27236] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/24/2018] [Indexed: 01/25/2023]
Abstract
The transient receptor potential melastatin-2 (TRPM2) channel belongs to the transient receptor potential channel superfamily and is a cation channel permeable to Na+ and Ca 2+ . The TRPM2 ion channel is expressed in the kidney and can be activated by various molecules such as hydrogen peroxide, calcium, and cyclic adenosine diphosphate (ADP)-ribose (cADPR) that are produced during acute kidney injury. In this study, we investigated the role of 8-bromo-cyclic ADP-ribose (8-Br-cADPR; a cADPR antagonist) in renal ischemia-reperfusion injury using biochemical and histopathological parameters. CD38, cADPR, tumor necrosis factor-α, interleukin-1β, and myeloperoxidase (inflammatory markers), urea and creatinine, hydrogen peroxide (oxidant), and catalase (antioxidant enzyme) levels that increase with ischemia-reperfusion injury decreased in the groups treated with 8-Br-cADPR. In addition, renin levels were elevated in the groups treated with 8-Br-cADPR. Histopathological examination revealed that 8-Br-cADPR reduced renal damage and the expression of caspase-3 and TRPM2. Our results suggest that the inhibition of TRPM2 ion channel may be a new treatment modality for ischemic acute kidney injury.
Collapse
Affiliation(s)
- Ersen Eraslan
- Department of Physiology, Faculty of Medicine, University of Bozok, Yozgat, Turkey
| | - Ayhan Tanyeli
- Department of Physiology, Faculty of Medicine, University of Atatürk, Erzurum, Turkey
| | - Elif Polat
- Department of Biochemistry, Faculty of Medicine, University of Atatürk, Erzurum, Turkey
| | - Elif Polat
- Department of Histology and Embryology, Faculty of Medicine, University of Namık Kemal, Tekirdağ, Turkey
| |
Collapse
|
11
|
The role of TRPM2 channels in neurons, glial cells and the blood-brain barrier in cerebral ischemia and hypoxia. Acta Pharmacol Sin 2018. [PMID: 29542681 PMCID: PMC5943904 DOI: 10.1038/aps.2017.194] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Stroke is one of the major causes of mortality and morbidity worldwide, yet novel therapeutic treatments for this condition are lacking. This review focuses on the roles of the transient receptor potential melastatin 2 (TRPM2) ion channels in cellular damage following hypoxia-ischemia and their potential as a future therapeutic target for stroke. Here, we highlight the complex molecular signaling that takes place in neurons, glial cells and the blood-brain barrier following ischemic insult. We also describe the evidence of TRPM2 involvement in these processes, as shown from numerous in vitro and in vivo studies that utilize genetic and pharmacological approaches. This evidence implicates TRPM2 in a broad range of pathways that take place every stage of cerebral ischemic injury, thus making TRPM2 a promising target for drug development for stroke and other neurodegenerative conditions of the central nervous system.
Collapse
|
12
|
Yamamoto S, Toda T, Yonezawa R, Negoro T, Shimizu S. Tyrphostin AG-related compounds attenuate H 2O 2-induced TRPM2-dependent and -independent cellular responses. J Pharmacol Sci 2017; 134:68-74. [PMID: 28532624 DOI: 10.1016/j.jphs.2017.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 03/22/2017] [Accepted: 04/28/2017] [Indexed: 01/13/2023] Open
Abstract
PURPOSE TRPM2 is a Ca2+-permeable channel that is activated by H2O2. TRPM2-mediated Ca2+ signaling has been implicated in the aggravation of inflammatory diseases. Therefore, the development of TRPM2 inhibitors to prevent the aggravation of these diseases is expected. We recently reported that some Tyrphostin AG-related compounds inhibited the H2O2-induced activation of TRPM2 by scavenging the intracellular hydroxyl radical. In the present study, we examined the effects of AG-related compounds on H2O2-induced cellular responses in human monocytic U937 cells, which functionally express TRPM2. METHODS The effects of AG-related compounds on H2O2-induced changes in intracellular Ca2+ concentrations, extracellular signal-regulated kinase (ERK) activation, and CXCL8 secretion were assessed using U937 cells. RESULTS Ca2+ influxes via TRPM2 in response to H2O2 were blocked by AG-related compounds. AG-related compounds also inhibited the H2O2-induced activation of ERK, and subsequent secretion of CXCL8 mediated by TRPM2-dependent and -independent mechanisms. CONCLUSION Our results show that AG-related compounds inhibit H2O2-induced CXCL8 secretion following ERK activation, which is mediated by TRPM2-dependent and -independent mechanisms in U937 cells. We previously reported that AG-related compounds blocked H2O2-induced TRPM2 activation by scavenging the hydroxyl radical. The inhibitory effects of AG-related compounds on TRPM2-independent responses may be due to scavenging of the hydroxyl radical.
Collapse
Affiliation(s)
- Shinichiro Yamamoto
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, 164-8530, Japan
| | - Takahiro Toda
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, 164-8530, Japan
| | - Ryo Yonezawa
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, 164-8530, Japan; Department of Hospital Pharmaceutics, Showa University School of Pharmacy, Tokyo, 142-8555, Japan
| | - Takaharu Negoro
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, 164-8530, Japan
| | - Shunichi Shimizu
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, 164-8530, Japan.
| |
Collapse
|
13
|
Markó L, Mannaa M, Haschler TN, Krämer S, Gollasch M. Renoprotection: focus on TRPV1, TRPV4, TRPC6 and TRPM2. Acta Physiol (Oxf) 2017; 219:589-612. [PMID: 28028935 DOI: 10.1111/apha.12828] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 04/22/2016] [Accepted: 10/31/2016] [Indexed: 01/09/2023]
Abstract
Members of the transient receptor potential (TRP) cation channel receptor family have unique sites of regulatory function in the kidney which enables them to promote regional vasodilatation and controlled Ca2+ influx into podocytes and tubular cells. Activated TRP vanilloid 1 receptor channels (TRPV1) have been found to elicit renoprotection in rodent models of acute kidney injury following ischaemia/reperfusion. Transient receptor potential cation channel, subfamily C, member 6 (TRPC6) in podocytes is involved in chronic proteinuric kidney disease, particularly in focal segmental glomerulosclerosis (FSGS). TRP vanilloid 4 receptor channels (TRPV4) are highly expressed in the kidney, where they induce Ca2+ influx into endothelial and tubular cells. TRP melastatin (TRPM2) non-selective cation channels are expressed in the cytoplasm and intracellular organelles, where their inhibition ameliorates ischaemic renal pathology. Although some of their basic properties have been recently identified, the renovascular role of TRPV1, TRPV4, TRPC6 and TRPM2 channels in disease states such as obesity, hypertension and diabetes is largely unknown. In this review, we discuss recent evidence for TRPV1, TRPV4, TRPC6 and TRPM2 serving as potential targets for acute and chronic renoprotection in chronic vascular and metabolic disease.
Collapse
Affiliation(s)
- L. Markó
- Experimental and Clinical Research Center; A Joint Cooperation Between the Charité Medical Faculty and the Max-Delbrück Center (MDC) for Molecular Medicine; Berlin Germany
| | - M. Mannaa
- Experimental and Clinical Research Center; A Joint Cooperation Between the Charité Medical Faculty and the Max-Delbrück Center (MDC) for Molecular Medicine; Berlin Germany
- Charité Campus Virchow; Nephrology/Intensive Care; Berlin Germany
- German Institute of Human Nutrition; Potsdam-Rehbrücke Germany
| | - T. N. Haschler
- Experimental and Clinical Research Center; A Joint Cooperation Between the Charité Medical Faculty and the Max-Delbrück Center (MDC) for Molecular Medicine; Berlin Germany
- German Institute of Human Nutrition; Potsdam-Rehbrücke Germany
| | - S. Krämer
- German Institute of Human Nutrition; Potsdam-Rehbrücke Germany
| | - M. Gollasch
- Experimental and Clinical Research Center; A Joint Cooperation Between the Charité Medical Faculty and the Max-Delbrück Center (MDC) for Molecular Medicine; Berlin Germany
- Charité Campus Virchow; Nephrology/Intensive Care; Berlin Germany
| |
Collapse
|
14
|
Bystander effects elicited by single-cell photo-oxidative blue-light stimulation in retinal pigment epithelium cell networks. Cell Death Discov 2017; 3:16071. [PMID: 28179989 PMCID: PMC5292780 DOI: 10.1038/cddiscovery.2016.71] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 07/26/2016] [Accepted: 08/19/2016] [Indexed: 02/07/2023] Open
Abstract
‘Bystander effect’ refers to the induction of biological effects in cells not directly targeted. The retinal pigment epithelium consists of hexagonal cells, forming a monolayer interconnected by gap junctions (GJs). Oxidative stress initiated in an individual cell by photostimulation (488 nm) triggered changes in reactive oxygen species (ROS), Ca2+ and mitochondrial membrane potential (ψm). The Ca2+ signal was transmitted to neighboring cells slowly and non-uniformly; the ROS signal spread fast and radially. Increased Ca2+ levels were associated with a loss in ψm. GJ blockers prevented the spreading of the Ca2+, but not the ROS-related signal. The GJ-mediated Ca2+ wave was associated with cell death by 24 h, requiring endoplasmic reticulum–mitochondria Ca2+ transfer. Ensuing cell death was correlated with baseline Ca2+ levels, and baseline Ca2+ levels were correlated with pigmentation. Hence, local oxidative stress in a donor cell can trigger changes in certain connected recipient cells, a signal that required GJ communication and an ROS-Ca2+ dual-hit. Finally, damage apparently occurred in susceptible cells, which correlated with baseline Ca2+ levels.
Collapse
|
15
|
Sun Y, Sukumaran P, Selvaraj S, Cilz NI, Schaar A, Lei S, Singh BB. TRPM2 Promotes Neurotoxin MPP +/MPTP-Induced Cell Death. Mol Neurobiol 2016; 55:409-420. [PMID: 27957685 DOI: 10.1007/s12035-016-0338-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 11/30/2016] [Indexed: 12/21/2022]
Abstract
In neurons, Ca2+ is essential for a variety of physiological processes that regulate gene transcription to neuronal growth and their survival. 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and 1-methyl-4-phenylpyridinium ions (MPP+) are potent neurotoxins that selectively destroys the dopaminergic (DA) neurons and mimics Parkinson's disease (PD) like symptoms, but the mechanism as how MPP+/MPTP effects DA neuron survival is not well-understood. In the present study, we found that MPP+ treatment increased the level of reactive oxygen species (ROS) that activates and upregulates the expression and function of melastatin-like transient receptor potential (TRPM) subfamily member, melastatin-like transient receptor potential channel 2 (TRPM2). Correspondingly, TRPM2 expression was also increased in substantia nigra of MPTP-induced PD mouse model and PD patients. ROS-mediated activation of TRPM2 resulted in an increased intracellular Ca2+, which in turn promoted cell death in SH-SY5Y cells. Intracellular Ca2+ overload caused by MPP+-induced ROS also affected calpain activity, followed by increased caspase 3 activities and activation of downstream apoptotic pathway. On the other hand, quenching of H2O2 by antioxidants, resveratrol (RSV), or N-acetylcysteine (NAC) effectively blocked TRPM2-mediated Ca2+ influx, decreased intracellular Ca2+ overload, and increased cell survival. Importantly, pharmacological inhibition of TRPM2 or knockdown of TRPM2 using siRNA, but not control siRNA, showed an increased protection by preventing MPP+-induced Ca2+ increase and inhibited apoptosis. Taken together, we show here a novel role for TRPM2 expression and function in MPP+-induced dopaminergic neuronal cell death.
Collapse
Affiliation(s)
- Yuyang Sun
- Department of Biomedical Science, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58201, USA
| | - Pramod Sukumaran
- Department of Biomedical Science, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58201, USA
| | - Senthil Selvaraj
- Department of Biomedical Science, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58201, USA
| | - Nicholas I Cilz
- Department of Biomedical Science, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58201, USA
| | - Anne Schaar
- Department of Biomedical Science, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58201, USA
| | - Saobo Lei
- Department of Biomedical Science, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58201, USA
| | - Brij B Singh
- Department of Biomedical Science, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58201, USA.
| |
Collapse
|
16
|
Yamamoto S, Shimizu S. Significance of TRP channels in oxidative stress. Eur J Pharmacol 2016; 793:109-111. [PMID: 27838397 DOI: 10.1016/j.ejphar.2016.11.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 10/20/2016] [Accepted: 11/02/2016] [Indexed: 01/06/2023]
Abstract
Reactive oxygen species induce oxidative stress, leading to cell damage, but also function as signal transduction molecules. Transient receptor potential (TRP) channels have been attracting increasing attention as Ca2+-permeable channels that sense environmental changes. The members of one class of TRP channels have emerged as reactive oxygen species sensors. The significance of Ca2+ signaling induced by the activation of reactive oxygen species-sensitive TRP channels under pathological conditions is currently being elucidated. The selective inhibition of reactive oxygen species-sensitive TRP channels represents a future challenge that may lead to new therapeutic strategies for the suppression of reactive oxygen species-related diseases.
Collapse
Affiliation(s)
- Shinichiro Yamamoto
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan
| | - Shunichi Shimizu
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan.
| |
Collapse
|
17
|
Targeting TRPM2 in ROS-Coupled Diseases. Pharmaceuticals (Basel) 2016; 9:ph9030057. [PMID: 27618067 PMCID: PMC5039510 DOI: 10.3390/ph9030057] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 08/05/2016] [Accepted: 09/05/2016] [Indexed: 12/15/2022] Open
Abstract
Under pathological conditions such as inflammation and ischemia-reperfusion injury large amounts of reactive oxygen species (ROS) are generated which, in return, contribute to the development and exacerbation of disease. The second member of the transient receptor potential (TRP) melastatin subfamily, TRPM2, is a Ca(2+)-permeable non-selective cation channel, activated by ROS in an ADP-ribose mediated fashion. In other words, TRPM2 functions as a transducer that converts oxidative stress into Ca(2+) signaling. There is good evidence that TRPM2 plays an important role in ROS-coupled diseases. For example, in monocytes the influx of Ca(2+) through TRPM2 activated by ROS contributes to the aggravation of inflammation via chemokine production. In this review, the focus is on TRPM2 as a molecular linker between ROS and Ca(2+) signaling in ROS-coupled diseases.
Collapse
|
18
|
Toda T, Yamamoto S, Yonezawa R, Mori Y, Shimizu S. Inhibitory effects of Tyrphostin AG-related compounds on oxidative stress-sensitive transient receptor potential channel activation. Eur J Pharmacol 2016; 786:19-28. [DOI: 10.1016/j.ejphar.2016.05.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/23/2016] [Accepted: 05/25/2016] [Indexed: 12/28/2022]
|
19
|
Chidgey J, Fraser PA, Aaronson PI. Reactive oxygen species facilitate the EDH response in arterioles by potentiating intracellular endothelial Ca(2+) release. Free Radic Biol Med 2016; 97:274-284. [PMID: 27320188 PMCID: PMC5005039 DOI: 10.1016/j.freeradbiomed.2016.06.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 06/14/2016] [Accepted: 06/15/2016] [Indexed: 11/17/2022]
Abstract
There is abundant evidence that H2O2 can act as an endothelium-derived hyperpolarizing factor in the resistance vasculature. However, whilst scavenging H2O2 can abolish endothelial dependent hyperpolarization (EDH) and the associated vascular relaxation in some arteries, EDH-dependent vasorelaxation can often be mimicked only by using relatively high concentrations of H2O2. We have examined the role of H2O2 in EDH-dependent vasodilatation by simultaneously measuring vascular diameter and changes in endothelial cell (EC) [Ca(2+)]i during the application of H2O2 or carbachol, which triggers EDH. Carbachol (10µM) induced dilatation of phenylephrine-preconstricted rat cremaster arterioles was largely (73%) preserved in the presence of indomethacin (3µM) and l-NAME (300µM). This residual NO- and prostacyclin-independent dilatation was reduced by 89% upon addition of apamin (0.5µM) and TRAM-34 (10µM), and by 74% when an extracellular ROS scavenging mixture of SOD and catalase (S&C; 100Uml(-1) each) was present. S&C also reduced the carbachol-induced EC [Ca(2+)]i increase by 74%. When applied in Ca(2+)-free external medium, carbachol caused a transient increase in EC [Ca(2+)]i. This was reduced by catalase, and was enhanced when 1µM H2O2 was present in the bath. H2O2 -induced dilatation, which occurred only at concentrations ≥100µM, was reduced by a blocking antibody to TRPM2, which had no effect on carbachol-induced responses. Similarly, iberotoxin and Rp-8bromo cGMP reduced the vasodilatation induced by H2O2, but not by carbachol. Inhibiting PLC, PLA2 or CYP450 2C9 each greatly reduced the carbachol-induced increase in EC [Ca(2+)]i and vasodilatation, but adding 10µM H2O2 during PLA2 or CYP450 2C9 inhibition completely restored both responses. The nature of the effective ROS species was investigated by using Fe(2+) chelators to block the formation of ∙OH. A cell permeant chelator was able to inhibit EC Ca(2+) store release, but cell impermeant chelators reduced both the vasodilatation and EC Ca(2+) influx, implying that ∙OH is required for these responses. The results indicate that rather than mediating EDH by acting directly on smooth muscle, H2O2 promotes EDH by acting within EC to enhance Ca(2+) release.
Collapse
Affiliation(s)
- James Chidgey
- King's College London, Faculty of Life Sciences and Medicine, Division of Asthma, Allergy & Lung Biology, London, United Kingdom
| | - Paul A Fraser
- King's College London, Faculty of Life Sciences and Medicine, Cardiovascular Division, London, United Kingdom
| | - Philip I Aaronson
- King's College London, Faculty of Life Sciences and Medicine, Division of Asthma, Allergy & Lung Biology, London, United Kingdom.
| |
Collapse
|
20
|
Yang Q, Lin TS, Burton C, Park SH, Ma Y. Physicochemical insights of irradiation-enhanced hydroxyl radical generation from ZnO nanoparticles. Toxicol Res (Camb) 2016; 5:482-491. [PMID: 30090362 PMCID: PMC6061809 DOI: 10.1039/c5tx00384a] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/21/2015] [Indexed: 11/21/2022] Open
Abstract
The widespread use of zinc oxide nanoparticles (ZnO NPs) has raised environmental and human health concerns owing to their significant cytotoxicity. Although their cytotoxic effects have been associated with reactive oxygen species (ROS), the physicochemical mechanism underlying this phenomenon remains poorly understood. In this study, the physicochemical properties of ZnO NPs were systematically investigated in relation to their effect on ROS generation. Factors that were found to affect hydroxyl radical (˙OH) generation included: NP concentration, irradiation, NP hydrodynamic size, localized pH, ionic strength, NP zeta-potential, and dissolved oxygen levels. The mechanism by which ˙OH was generated under alkaline conditions was found to obey first-order reaction kinetics that followed the conversion of OH- anions and dissolved O2 to ˙OH. Based on these findings, we propose that ZnO NP cytotoxicity involves ˙OH adsorption to the nanoparticle surface, creating a highly localized source of ROS capable of potentiating oxidative damage to cellular structures. This hypothesis was evaluated with time-resolved intracellular calcium [Ca] i imaging that irradiated ZnO NPs triggered cytoplasmic calcium influxes and facilitated nuclear degradation. Together these findings present a novel physicochemical mechanism for ˙OH generation from ZnO NPs with significant implications for nanoparticle cytotoxicity and their relation to human health.
Collapse
Affiliation(s)
- Qingbo Yang
- Department of Chemistry and Center for Single Nanoparticle , Single Cell , and Single Molecule Monitoring , Missouri University of Science and Technology , Rolla , MO 65409 , USA . ; ; Tel: +1-573-341-6220
| | - Tien-Sung Lin
- Department of Chemistry , Washington University in St. Louis , St. Louis , MO 63130 , USA
| | - Casey Burton
- Department of Chemistry and Center for Single Nanoparticle , Single Cell , and Single Molecule Monitoring , Missouri University of Science and Technology , Rolla , MO 65409 , USA . ; ; Tel: +1-573-341-6220
| | - Sung-Ho Park
- Department of Chemistry , Washington University in St. Louis , St. Louis , MO 63130 , USA
| | - Yinfa Ma
- Department of Chemistry and Center for Single Nanoparticle , Single Cell , and Single Molecule Monitoring , Missouri University of Science and Technology , Rolla , MO 65409 , USA . ; ; Tel: +1-573-341-6220
| |
Collapse
|
21
|
Iron stimulates plasma-activated medium-induced A549 cell injury. Sci Rep 2016; 6:20928. [PMID: 26865334 PMCID: PMC4750041 DOI: 10.1038/srep20928] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 01/13/2016] [Indexed: 01/20/2023] Open
Abstract
Non-thermal atmospheric pressure plasma is applicable to living cells and has emerged as a novel technology for cancer therapy. Plasma has recently been shown to affect cells not only by direct irradiation, but also by indirect treatments with previously prepared plasma-activated medium (PAM). Iron is an indispensable element but is also potentially toxic because it generates the hydroxyl radical (•OH) in the presence of hydrogen peroxide (H2O2) via the Fenton reaction. The aim of the present study was to demonstrate the contribution of iron to PAM-induced A549 adenocarcinoma cell apoptosis. We detected the generation of •OH and elevation of intracellular ferrous ions in PAM-treated cells and found that they were inhibited by iron chelator. The elevations observed in ferrous ions may have been due to their release from the intracellular iron store, ferritin. Hydroxyl radical-induced DNA injury was followed by the activation of poly(ADP-ribose) polymerase-1, depletion of NAD+ and ATP, and elevations in intracellular Ca2+. The sensitivities of normal cells such as smooth muscle cells and keratinocytes to PAM were less than that of A549 cells. These results demonstrated that H2O2 in PAM and/or •OH generated in the presence of iron ions disturbed the mitochondrial-nuclear network in cancer cells.
Collapse
|
22
|
Sfaxi I, Charradi K, Limam F, El May MV, Aouani E. Grape seed and skin extract protects against arsenic trioxide induced oxidative stress in rat heart. Can J Physiol Pharmacol 2016; 94:168-176. [DOI: 10.1139/cjpp-2015-0088] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Arsenic is a metalloid found in water, soil, and air from natural and anthropogenic sources, and is commonly found in inorganic as well as organic forms. The clinical use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL) is limited by its cardiotoxic side effects. Grape seed and skin extract (GSSE) is a polyphenolic mixture with antioxidant properties. This study aimed to evaluate the protective effect of GSSE on arsenic-induced cardiac oxidative stress and injury. Animals exposed to 2.5 mg/kg As2O3 for 21 days exhibited a relevant increase in heart lipoperoxidation, protein carbonylation, and inflammation, as well as a drop in the activity of antioxidant enzymes such as catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GPx). In addition, As2O3 disturbed heart lipidemia and lipase activity, transition metals distribution and the associated enzymes, intracellular mediators such as calcium and the associated calpain activity, as well as myocardial architecture. Treatment with 4 g/kg GSSE protected against most of the deleterious effects provoked by As2O3. Our data suggest that GSSE has the potential to protect against As2O3-induced cardiotoxicity.
Collapse
Affiliation(s)
- Ichraf Sfaxi
- Laboratoire des Substances Bioactives (LSBA), Centre de Biotechnologie de Borj Cedria, BP-901, 2050 Hammam-Lif, Tunisie
- Université de Carthage, Faculté des Sciences de Bizerte, 7021 Jarzouna, Tunisie
| | - Kamel Charradi
- Laboratoire des Substances Bioactives (LSBA), Centre de Biotechnologie de Borj Cedria, BP-901, 2050 Hammam-Lif, Tunisie
- Université de Carthage, Faculté des Sciences de Bizerte, 7021 Jarzouna, Tunisie
| | - Ferid Limam
- Laboratoire des Substances Bioactives (LSBA), Centre de Biotechnologie de Borj Cedria, BP-901, 2050 Hammam-Lif, Tunisie
| | | | - Ezzedine Aouani
- Laboratoire des Substances Bioactives (LSBA), Centre de Biotechnologie de Borj Cedria, BP-901, 2050 Hammam-Lif, Tunisie
- Université de Carthage, Faculté des Sciences de Bizerte, 7021 Jarzouna, Tunisie
| |
Collapse
|
23
|
Shimizu S, Yonezawa R, Negoro T, Yamamoto S, Numata T, Ishii M, Mori Y, Toda T. Sensitization of H2O2-induced TRPM2 activation and subsequent interleukin-8 (CXCL8) production by intracellular Fe(2+) in human monocytic U937 cells. Int J Biochem Cell Biol 2015; 68:119-27. [PMID: 26386353 DOI: 10.1016/j.biocel.2015.09.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 09/07/2015] [Accepted: 09/14/2015] [Indexed: 01/16/2023]
Abstract
Transient receptor potential melastatin 2 (TRPM2) is an oxidative stress-sensitive Ca(2+)-permeable channel. In monocytes/macrophages, H2O2-induced TRPM2 activation causes cell death and/or production of chemokines that aggravate inflammatory diseases. However, relatively high concentrations of H2O2 are required for activation of TRPM2 channels in vitro. Thus, in the present study, factors that sensitize TRPM2 channels to H2O2 were identified and subsequent physiological responses were examined in U937 human monocytes. Temperature increase from 30°C to 37°C enhanced H2O2-induced TRPM2-mediated increase in intracellular free Ca(2+) ([Ca(2+)]i) in TRPM2-expressing HEK 293 cells (TRPM2/HEK cells). The H2O2-induced TRPM2 activation enhanced by the higher temperature was dramatically sensitized by intracellular Fe(2+)-accumulation following pretreatment with FeSO4. Thus intracellular Fe(2+)-accumulation sensitizes H2O2-induced TRPM2 activation at around body temperature. Moreover, intracellular Fe(2+)-accumulation increased poly(ADP-ribose) levels in nuclei by H2O2 treatment, and the sensitization of H2O2-induced TRPM2 activation were almost completely blocked by poly(ADP-ribose) polymerase inhibitors, suggesting that intracellular Fe(2+)-accumulation enhances H2O2-induced TRPM2 activation by increase of ADP-ribose production through poly(ADP-ribose) polymerase pathway. Similarly, pretreatment with FeSO4 stimulated H2O2-induced TRPM2 activation at 37°C in U937 cells and enhanced H2O2-induced ERK phosphorylation and interleukin-8 (CXCL8) production. Although the addition of H2O2 to cells under conditions of intracellular Fe(2+)-accumulation caused cell death, concentration of H2O2 required for CXCL8 production was lower than that resulting in cell death. These results indicate that intracellular Fe(2+)-accumulation sensitizes TRPM2 channels to H2O2 and subsequently produces CXCL8 at around body temperature. It is possible that sensitization of H2O2-induced TRPM2 channels by Fe(2+) may implicated in hemorrhagic brain injury via aggravation of inflammation, since Fe(2+) is released by heme degradation under intracerebral hemorrhage.
Collapse
Affiliation(s)
- Shunichi Shimizu
- Laboratory of Pharmacology, Department of Clinical Pharmacy, Yokohama College of Pharmacy, Yokohama 245-0066, Japan; Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo 164-8530, Japan.
| | - Ryo Yonezawa
- Division of Physiology and Pathology, Department of Pharmacology, Toxicology and Therapeutics, Showa University School of Pharmacy, Tokyo 142-8555, Japan
| | - Takaharu Negoro
- Department of Pharmacogenomics, Showa University School of Pharmacy, Tokyo 142-8555, Japan
| | - Shinichiro Yamamoto
- Department of Molecular Cell Biology and Medicine, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima 770-8505, Japan
| | - Tomohiro Numata
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Masakazu Ishii
- Division of Physiology and Pathology, Department of Pharmacology, Toxicology and Therapeutics, Showa University School of Pharmacy, Tokyo 142-8555, Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Takahiro Toda
- Laboratory of Pharmacology, Department of Clinical Pharmacy, Yokohama College of Pharmacy, Yokohama 245-0066, Japan
| |
Collapse
|
24
|
Veit F, Pak O, Brandes RP, Weissmann N. Hypoxia-dependent reactive oxygen species signaling in the pulmonary circulation: focus on ion channels. Antioxid Redox Signal 2015; 22:537-52. [PMID: 25545236 PMCID: PMC4322788 DOI: 10.1089/ars.2014.6234] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE An acute lack of oxygen in the lung causes hypoxic pulmonary vasoconstriction, which optimizes gas exchange. In contrast, chronic hypoxia triggers a pathological vascular remodeling causing pulmonary hypertension, and ischemia can cause vascular damage culminating in lung edema. RECENT ADVANCES Regulation of ion channel expression and gating by cellular redox state is a widely accepted mechanism; however, it remains a matter of debate whether an increase or a decrease in reactive oxygen species (ROS) occurs under hypoxic conditions. Ion channel redox regulation has been described in detail for some ion channels, such as Kv channels or TRPC6. However, in general, information on ion channel redox regulation remains scant. CRITICAL ISSUES AND FUTURE DIRECTIONS In addition to the debate of increased versus decreased ROS production during hypoxia, we aim here at describing and deciphering why different oxidants, under different conditions, can cause both activation and inhibition of channel activity. While the upstream pathways affecting channel gating are often well described, we need a better understanding of redox protein modifications to be able to determine the complexity of ion channel redox regulation. Against this background, we summarize the current knowledge on hypoxia-induced ROS-mediated ion channel signaling in the pulmonary circulation.
Collapse
Affiliation(s)
- Florian Veit
- 1 Excellence Cluster Cardiopulmonary System (ECCPS), Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL) , Giessen, Germany
| | | | | | | |
Collapse
|
25
|
Inhibitory effects of AG490 on H2O2-induced TRPM2-mediated Ca2+ entry. Eur J Pharmacol 2014; 742:22-30. [DOI: 10.1016/j.ejphar.2014.08.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 08/19/2014] [Accepted: 08/20/2014] [Indexed: 01/12/2023]
|
26
|
Ishii M, Hagiwara T, Mori Y, Shimizu S. Involvement of TRPM2 and L-type Ca²⁺ channels in Ca²⁺ entry and cell death induced by hydrogen peroxide in rat β-cell line RIN-5F. J Toxicol Sci 2014; 39:199-209. [PMID: 24646700 DOI: 10.2131/jts.39.199] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Ca²⁺ overload is one of the mechanisms for H₂O₂-induced cell death in rat pancreatic β-cell line RIN-5F cells. RIN-5F cells express TRPM2, which is a Ca²⁺-permeable channel activated by H₂O₂, and voltage-dependent L-type Ca²⁺ channels, both of which induce Ca²⁺ entry by H₂O₂. This study examined the contribution of these channels to H₂O₂-induced Ca²⁺ entry and cell death in RIN-5F cells. Cytosolic Ca²⁺ concentration was measured using fura-2 as a Ca²⁺ indicator. Cell death was estimated by trypan blue exclusion. Pre-treatment with poly(ADP-ribose) polymerase (PARP) inhibitors, which inhibit TRPM2 activation, strongly reduced Ca²⁺ entry by H₂O₂. The PARP inhibitors used had no effect on the Ca²⁺ elevation by voltage-dependent L-type Ca²⁺ channels. On the other hand, pre-treatment with L-type Ca²⁺ channel blockers, which did not affect TRPM2 activation, partly reduced H₂O₂-induced Ca²⁺ entry. Treatment with PARP inhibitors but not L-type Ca²⁺ channel blockers, around the early phase in H₂O₂-induced Ca²⁺ elevation, also reduced the late phase. Moreover, H₂O₂-induced RIN-5F cell death was strongly attenuated by PARP inhibitors, in compared to L-type Ca²⁺ channel blockers. Our results suggest that TRPM2 channels rather than L-type Ca²⁺ channels primarily contribute to H₂O₂-induced Ca²⁺ entry and cell death.
Collapse
Affiliation(s)
- Masakazu Ishii
- Department of Pharmacology, Toxicology and Therapeutics, Division of Physiology and Pathology, Showa University School of Pharmacy
| | | | | | | |
Collapse
|
27
|
Abstract
SIGNIFICANCE Environmental and endogenous reactive species such as reactive oxygen species (ROS), reactive nitrogen species (RNS), and other electrophiles are not only known to exert toxic effects on organisms, but are also emerging as molecules that mediate cell signaling responses. However, the mechanisms underlying this cellular redox signaling by reactive species remains largely uncharacterized. RECENT ADVANCES Ca2+-permeable cation channels encoded by the transient receptor potential (trp) gene superfamily are characterized by a wide variety of activation triggers that act from outside and inside the cell. Recent studies have revealed that multiple TRP channels sense reactive species and induce diverse physiological and pathological responses, such as cell death, chemokine production, and pain transduction. TRP channels sense reactive species either indirectly through second messengers or directly via oxidative modification of cysteine residues. In this review, we describe the activation mechanisms and biological roles of redox-sensitive TRP channels, including TRPM2, TRPM7, TRPC5, TRPV1, and TRPA1. CRITICAL ISSUES The sensitivity of TRP channels to reactive species in vitro has been well characterized using molecular and pharmacological approaches. However, the precise activation mechanism(s) and in vivo function(s) of ROS/RNS-sensitive TRP channels remain elusive. FUTURE DIRECTIONS Redox sensitivity of TRP channels has been shown to mediate previously unexplained biological phenomena and is involved in various pathologies. Understanding the physiological significance and activation mechanisms of TRP channel regulation by reactive species may lead to TRP channels becoming viable pharmacological targets, and modulators of these channels may offer therapeutic options for previously untreatable diseases.
Collapse
Affiliation(s)
- Daisuke Kozai
- 1 Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University , Kyoto, Japan
| | | | | |
Collapse
|
28
|
de Paula RG, Ornelas AMDM, Morais ER, Borges WDC, Natale M, Magalhães LG, Rodrigues V. Biochemical characterization and role of the proteasome in the oxidative stress response of adult Schistosoma mansoni worms. Parasitol Res 2014; 113:2887-97. [PMID: 24870249 DOI: 10.1007/s00436-014-3950-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 05/06/2014] [Indexed: 11/28/2022]
Abstract
The trematode Schistosoma mansoni, an important parasite of humans, is the principle agent of the disease schistosomiasis. In the human host, one of the most important stress factors of this parasite is the oxidative stress generated by both the metabolism of the worm and the immune system of the host. The proteasomal system is responsible for protein homeostasis during oxidative stress. The 26S proteasome is a multicatalytic protease formed by two compartments, a 20S core and regulatory particle 19S, and controls the degradation of intracellular proteins, hence regulating many cellular processes. In the present report, we describe the biochemical characterization and role of the 20S proteasome in the response of adult S. mansoni worms exposed to hydrogen peroxide. Characterization of the response to the oxidative stress included the evaluation of viability, egg production, mortality, tegument integrity, and both expression and activity of proteasome. We observed decreases in viability, egg production as well as 100% mortality at the higher concentrations of hydrogen peroxide tested. The main changes observed in the tegument of adult worms were peeling as well as the appearance of bubbles and a decrease of spines on the tubercles. Furthermore, there were increases in 26S activity to the same extent as 20S proteasome activity, although there was increase of 20S proteasome content, suggesting that degradation of protein oxidized in adult worms is due to the 20S proteasome. It was demonstrated that adult S. mansoni worms are sensitive to oxidative stress, and that a variety of processes in this parasite are altered under this condition. The work contributes to a better understanding of the mechanisms employed by S. mansoni to survive under oxidative stress.
Collapse
Affiliation(s)
- Renato Graciano de Paula
- Faculdade de Medicina de Ribeirão Preto, Departamento de Bioquímica e Imunologia, Universidade de São Paulo, Avenida Bandeirantes, 3900, 14049-900, Ribeirão Preto, SP, Brazil,
| | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Acetaminophen (paracetamol) is the most frequently used analgesic and antipyretic drug available over the counter. At the same time, acetaminophen overdose is the most common cause of acute liver failure and the leading cause of chronic liver damage requiring liver transplantation in developed countries. Acetaminophen overdose causes a multitude of interrelated biochemical reactions in hepatocytes including the formation of reactive oxygen species, deregulation of Ca(2+) homeostasis, covalent modification and oxidation of proteins, lipid peroxidation, and DNA fragmentation. Although an increase in intracellular Ca(2+) concentration in hepatocytes is a known consequence of acetaminophen overdose, its importance in acetaminophen-induced liver toxicity is not well understood, primarily due to lack of knowledge about the source of the Ca(2+) rise. Here we report that the channel responsible for Ca(2+) entry in hepatocytes in acetaminophen overdose is the Transient Receptor Potential Melanostatine 2 (TRPM2) cation channel. We show by whole-cell patch clamping that treatment of hepatocytes with acetaminophen results in activation of a cation current similar to that activated by H2O2 or the intracellular application of ADP ribose. siRNA-mediated knockdown of TRPM2 in hepatocytes inhibits activation of the current by either acetaminophen or H2O2. In TRPM2 knockout mice, acetaminophen-induced liver damage, assessed by the blood concentration of liver enzymes and liver histology, is significantly diminished compared with wild-type mice. The presented data strongly suggest that TRPM2 channels are essential in the mechanism of acetaminophen-induced hepatocellular death.
Collapse
|
30
|
Abstract
TRPM2 (transient receptor potential melastatin 2) is a non-selective Ca2+-permeable cation channel activated by ADPR (adenosine diphosphoribose) and H2O2. It is widely expressed in mammalian cells and plays an important role in the regulation of various cell functions. However, the mechanisms of TRPM2 channel activation are not fully understood. Previously, we reported that TRPM2 channel activation is induced by high intracellular Cl- concentration. In the present study, we investigated the functional role of Lys1110 in the membrane-proximal C-terminal region by site-directed mutagenesis. Replacement of the positively charged amino acid lysine (Lys1110) with the neutrally charged amino acid asparagine (K1110N) or the negatively charged amino acid glutamic acid (K1110E) generated mutants that failed to induce an increase in free cytosolic calcium concentration ([Ca2+]i) not only by intracellular injection of Cl-, but also by H2O2 or ADPR. However, a mutant generated by replacing the lysine residue with a positively charged amino acid arginine (K1110R) displayed channel activity similar to wild-type TRPM2. Interestingly, in the K1107N/K1110N double-point mutant, the impaired function of the K1110N mutant in response to ADPR and H2O2, but not to Cl-, was recovered. There were no changes in protein expression, membrane trafficking and oligomerization of the mutant channels. The extent of [Ca2+]i increase by H2O2 in HEK (human embryonic kidney)-293 cells expressing TRPM2 mutants was well correlated with the degree of susceptibility to H2O2-induced cell death. These results display the crucial role of a positively charged amino acid residue at position 1110 for TRPM2 channel activity.
Collapse
|
31
|
Abstract
TRPM2 is the second member of the transient receptor potential melastatin-related (TRPM) family of cation channels. The protein is widely expressed including in the brain, immune system, endocrine cells, and endothelia. It embodies both ion channel functionality and enzymatic ADP-ribose (ADPr) hydrolase activity. TRPM2 is a Ca(2+)-permeable nonselective cation channel embedded in the plasma membrane and/or lysosomal compartments that is primarily activated in a synergistic fashion by intracellular ADP-ribose (ADPr) and Ca(2+). It is also activated by reactive oxygen and nitrogen species (ROS/NOS) and enhanced by additional factors, such as cyclic ADPr and NAADP, while inhibited by permeating protons (acidic pH) and adenosine monophosphate (AMP). Activation of TRPM2 leads to increases in intracellular Ca(2+) levels, which can serve signaling roles in inflammatory and secretory cells through release of vesicular mediators (e.g., cytokines, neurotransmitters, insulin) and in extreme cases can induce apoptotic and necrotic cell death under oxidative stress.
Collapse
Affiliation(s)
- Malika Faouzi
- Center for Biomedical Research, The Queen's Medical Center, 1301 Punchbowl Street, Honolulu, HI, 96813, USA,
| | | |
Collapse
|
32
|
Hiriart M, Velasco M, Larqué C, Diaz-Garcia CM. Metabolic Syndrome and Ionic Channels in Pancreatic Beta Cells. THE PANCREATIC BETA CELL 2014; 95:87-114. [DOI: 10.1016/b978-0-12-800174-5.00004-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
33
|
Hecquet CM, Zhang M, Mittal M, Vogel SM, Di A, Gao X, Bonini MG, Malik AB. Cooperative interaction of trp melastatin channel transient receptor potential (TRPM2) with its splice variant TRPM2 short variant is essential for endothelial cell apoptosis. Circ Res 2013; 114:469-79. [PMID: 24337049 DOI: 10.1161/circresaha.114.302414] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Oxidants generated by activated endothelial cells are known to induce apoptosis, a pathogenic feature of vascular injury and inflammation from multiple pathogeneses. The melastatin-family transient receptor potential 2 (TRPM2) channel is an oxidant-sensitive Ca2+ permeable channel implicated in mediating apoptosis; however, the mechanisms of gating of the supranormal Ca2+ influx required for initiating of apoptosis are not understood. OBJECTIVE Here, we addressed the role of TRPM2 and its interaction with the short splice variant TRPM2 short variant (TRPM2-S) in mediating the Ca2+ entry burst required for induction of endothelial cell apoptosis. METHODS AND RESULTS We observed that TRPM2-S was basally associated with TRPM2 in the endothelial plasmalemma, and this interaction functioned to suppress TRPM2-dependent Ca2+ gating constitutively. Reactive oxygen species production in endothelial cells or directly applying reactive oxygen species induced protein kinase C-α activation and phosphorylation of TRPM2 at Ser 39. This in turn stimulated a large entry of Ca2+ and activated the apoptosis pathway. A similar TRPM2-dependent endothelial apoptosis mechanism was seen in intact vessels. The protein kinase C-α-activated phosphoswitch opened the TRPM2 channel to allow large Ca2+ influx by releasing TRPM2-S inhibition of TRPM2, which in turn activated caspase-3 and cleaved the caspase substrate poly(ADP-ribose) polymerase. CONCLUSIONS Here, we describe a fundamental mechanism by which activation of the trp superfamily TRPM2 channel induces apoptosis of endothelial cells. The signaling mechanism involves reactive oxygen species-induced protein kinase C-α activation resulting in phosphorylation of TRPM2-S that allows enhanced TRPM2-mediated gating of Ca2+ and activation of the apoptosis program. Strategies aimed at preventing the uncoupling of TRPM2-S from TRPM2 and subsequent Ca2+ gating during oxidative stress may mitigate endothelial apoptosis and its consequences in mediating vascular injury and inflammation.
Collapse
Affiliation(s)
- Claudie M Hecquet
- From the Department of Pharmacology and the Center for Lung and Vascular Biology (C.M.H., M.Z., M.M., S.M.V., A.D., X.G., M.G.B., A.B.M.) and Section of Cardiology (M.G.B.), College of Medicine, University of Illinois, Chicago
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Nazıroğlu M, Özgül C. Vitamin E modulates oxidative stress and protein kinase C activator (PMA)-induced TRPM2 channel gate in dorsal root ganglion of rats. J Bioenerg Biomembr 2013; 45:541-9. [PMID: 23943124 DOI: 10.1007/s10863-013-9524-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 07/30/2013] [Indexed: 01/07/2023]
Abstract
It is well known that Ca(2+) influx through cation channels induces peripheral pain in dorsal root ganglion (DRG) neurons. Melastatin-like transient receptor potential 2 (TRPM2) channel is a oxidative redox sensitive Ca(2+)-permeable cation channel. There is scarce report on block of the channels. Since the mechanisms that lead to TRPM2 inhibition in response to oxidative stress and protein kinase C (PKC) activation are not understood, we investigated effects of the antioxidants on the inhibition of TRPM2 channel currents in the DRG neurons of rats. The DRG peripheral neurons were freshly isolated from rats and the neurons were incubated by phorbol 12-myristate 13-acetate (PMA) which leads to activation of PKC and cause oxidative stress. In whole-cell patch clamp experiments, TRPM2 currents in the DRG incubated with PMA were stimulated by H2O2. In addition, the PMA-induced activation of TRPM2 channels were blocked by nonspecific TRPM2 channels inhibitors [2-aminoethyl diphenylborinate (2-APB) and N-(p-amylcinnamoyl)anthranilic acid (ACA)]. The currents in the neurons are also totally blocked by vitamin E incubation. However, administration of catalase and vitamin C with/without the vitamin E incubation did not block the currents. In conclusion, we indicated that vitamin E modulated oxidative stress-induced TRPM2 channel activation in the DRG neurons. The results may be useful modulation of oxidative stress-induced peripheral pain in sensory neurons.
Collapse
Affiliation(s)
- Mustafa Nazıroğlu
- Neuroscience Research Center, University of Suleyman Demirel, Isparta, Turkey,
| | | |
Collapse
|
35
|
Oda S, Uchida K, Wang X, Lee J, Shimada Y, Tominaga M, Kadowaki M. TRPM2 contributes to antigen-stimulated Ca²⁺ influx in mucosal mast cells. Pflugers Arch 2013; 465:1023-30. [PMID: 23371039 DOI: 10.1007/s00424-013-1219-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Revised: 01/10/2013] [Accepted: 01/10/2013] [Indexed: 11/29/2022]
Abstract
Food allergy (FA) is a common allergic disease without any currently available effective drug therapies. Mucosal mast cells (MMCs) play a particularly important role in FA, and the increase in their cytosolic Ca(2+) concentration ([Ca(2+)]cyt) is considered to be a principal component of the degranulation process. However, the mechanisms governing Ca(2+) influx remain poorly understood in MMCs. Recent reports have highlighted the functions of the transient receptor potential melastatin 2 (TRPM2) channel in immunocytes, including its role in monocyte chemokine production and macrophage phagocytic activity. Although TRPM2 gene expression has been demonstrated in mast cells, the significance of such expression remains virtually unknown. In this study, we found that antigen-stimulated degranulation was significantly reduced in mucosal-type bone marrow-derived mast cells (mBMMCs) prepared from TRPM2-knockout (TRPM2-KO) mice (TRPM2-KO mBMMCs) and was suppressed following the administration of three TRPM2 inhibitors with different chemical structures, including econazole, flufenamic acid (FFA), and 2-aminoethoxydiphenyl borate. Furthermore, the antigen-stimulated increase in [Ca(2+)]cyt was significantly decreased in TRPM2-KO mBMMCs and was also suppressed by the TRPM2 inhibitors econazole and FFA. In addition, thapsigargin-induced increase in [Ca(2+)]cyt was significantly decreased in TRPM2-KO mBMMCs. These results suggest that TRPM2 may participate in antigen-induced extracellular Ca(2+) influx and subsequent degranulation. In addition, TRPM2 inhibitors were shown to improve food allergic reactions in a mouse model. Together, these results suggest that TRPM2 inhibitors suppress MMC degranulation via regulation of the increase in [Ca(2+)]cyt. Thus, TRPM2 may play a key role in degranulation by modulating intracellular Ca(2+) in MMCs.
Collapse
Affiliation(s)
- Satoshi Oda
- Division of Gastrointestinal Pathophysiology, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | | | | | | | | | | | | |
Collapse
|
36
|
Zeng B, Chen GL, Xu SZ. Divalent copper is a potent extracellular blocker for TRPM2 channel. Biochem Biophys Res Commun 2012; 424:279-84. [PMID: 22750002 DOI: 10.1016/j.bbrc.2012.06.107] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 06/20/2012] [Indexed: 10/28/2022]
Abstract
Transient receptor potential melastatin 2 (TRPM2) is a Ca(2+)-permeable cationic channel in the TRP channel family. The channel activity can be regulated by reactive oxygen species (ROS) and cellular acidification, which has been implicated to the pathogenesis of diabetes and some neuronal disorders. However, little is known about the effect of redox-active metal ions, such as copper, on TRPM2 channels. Here we investigated the effect of divalent copper on TRPM2. TRPM2 channel was over-expressed in HEK-293 cells and the whole-cell current was recorded by patch clamp. We found the whole-cell current evoked by intracellular ADP-ribose was potently inhibited by Cu(2+) with a half maximal inhibitory concentration (IC(50)) of 2.59 μM. The inhibitory effect was irreversible. The single channel activity was abolished in the outside-out patches, and intracellular application of Cu(2+) did not prevent the channel activation, suggesting that the action site of Cu(2+) is located in the extracellular domains of the channel. TRPM2 current was also blocked by Hg(2+), Pb(2+), Fe(2+) and Se(2+). We concluded that Cu(2+) is a potent TRPM2 channel blocker. The sensitivity of TRPM2 channel to heavy metal ions could be a new mechanism for the pathogenesis of some metal ion-related diseases.
Collapse
Affiliation(s)
- Bo Zeng
- Centre for Cardiovascular and Metabolic Research, Hull York Medical School, University Hull, Hull, HU6 7RX, UK
| | | | | |
Collapse
|
37
|
Cao DS, Zhong L, Hsieh TH, Abooj M, Bishnoi M, Hughes L, Premkumar LS. Expression of transient receptor potential ankyrin 1 (TRPA1) and its role in insulin release from rat pancreatic beta cells. PLoS One 2012; 7:e38005. [PMID: 22701540 PMCID: PMC3365106 DOI: 10.1371/journal.pone.0038005] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 05/01/2012] [Indexed: 11/19/2022] Open
Abstract
Objective Several transient receptor potential (TRP) channels are expressed in pancreatic beta cells and have been proposed to be involved in insulin secretion. However, the endogenous ligands for these channels are far from clear. Here, we demonstrate the expression of the transient receptor potential ankyrin 1 (TRPA1) ion channel in the pancreatic beta cells and its role in insulin release. TRPA1 is an attractive candidate for inducing insulin release because it is calcium permeable and is activated by molecules that are produced during oxidative glycolysis. Methods Immunohistochemistry, RT-PCR, and Western blot techniques were used to determine the expression of TRPA1 channel. Ca2+ fluorescence imaging and electrophysiology (voltage- and current-clamp) techniques were used to study the channel properties. TRPA1-mediated insulin release was determined using ELISA. Results TRPA1 is abundantly expressed in a rat pancreatic beta cell line and freshly isolated rat pancreatic beta cells, but not in pancreatic alpha cells. Activation of TRPA1 by allyl isothiocyanate (AITC), hydrogen peroxide (H2O2), 4-hydroxynonenal (4-HNE), and cyclopentenone prostaglandins (PGJ2) and a novel agonist methylglyoxal (MG) induces membrane current, depolarization, and Ca2+ influx leading to generation of action potentials in a pancreatic beta cell line and primary cultured pancreatic beta cells. Activation of TRPA1 by agonists stimulates insulin release in pancreatic beta cells that can be inhibited by TRPA1 antagonists such as HC030031 or AP-18 and by RNA interference. TRPA1-mediated insulin release is also observed in conditions of voltage-gated Na+ and Ca2+ channel blockade as well as ATP sensitive potassium (KATP) channel activation. Conclusions We propose that endogenous and exogenous ligands of TRPA1 cause Ca2+ influx and induce basal insulin release and that TRPA1-mediated depolarization acts synergistically with KATP channel blockade to facilitate insulin release.
Collapse
Affiliation(s)
- De-Shou Cao
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
- * E-mail: (DSC); (LSP)
| | - Linlin Zhong
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
| | - Tsung-han Hsieh
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
| | - Mruvil Abooj
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
| | - Mahendra Bishnoi
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
| | - Lauren Hughes
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
| | - Louis S. Premkumar
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, United States of America
- * E-mail: (DSC); (LSP)
| |
Collapse
|
38
|
TRPM2 channel protective properties of N-acetylcysteine on cytosolic glutathione depletion dependent oxidative stress and Ca2+ influx in rat dorsal root ganglion. Physiol Behav 2012; 106:122-8. [DOI: 10.1016/j.physbeh.2012.01.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 01/11/2012] [Accepted: 01/17/2012] [Indexed: 01/05/2023]
|
39
|
Numazawa S, Takase M, Ahiko T, Ishii M, Shimizu SI, Yoshida T. Possible Involvement of Transient Receptor Potential Channels in Electrophile-Induced Insulin Secretion from RINm5F Cells. Biol Pharm Bull 2012; 35:346-54. [DOI: 10.1248/bpb.35.346] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Satoshi Numazawa
- Department of Biochemical Toxicology, Showa University School of Pharmacy
| | - Makiko Takase
- Department of Biochemical Toxicology, Showa University School of Pharmacy
| | - Tomomi Ahiko
- Department of Biochemical Toxicology, Showa University School of Pharmacy
| | - Masakazu Ishii
- Department of Pathophysiology, Showa University School of Pharmacy
| | | | - Takemi Yoshida
- Department of Biochemical Toxicology, Showa University School of Pharmacy
| |
Collapse
|
40
|
Takada M, Noguchi A, Sayama Y, Kurohane Kaneko Y, Ishikawa T. Inositol 1,4,5-trisphosphate receptor-mediated initial Ca(2+) mobilization constitutes a triggering signal for hydrogen peroxide-induced apoptosis in INS-1 β-cells. Biol Pharm Bull 2011; 34:954-8. [PMID: 21719997 DOI: 10.1248/bpb.34.954] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Reactive oxygen species, including hydrogen peroxide (H(2)O(2)), are known to induce β-cell apoptosis. The present study investigated the role of Ca(2+) in H(2)O(2)-induced apoptosis of the β-cell line INS-1. Annexin V assay with flow cytometry and DNA ladder assay demonstrated that treatment of INS-1 cells with 100 µM H(2)O(2) for 18 h significantly increased apoptotic cells. A comparable level of apoptosis was also observed after 18 h when the cells were treated with 100 µM H(2)O(2) only for initial 30 min. The H(2)O(2)-induced apoptosis was abolished by 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetra(acetoxymethyl)ester (BAPTA/AM), a chelator of intracellular Ca(2+), by 2-aminoethoxydiphenylborate (2-APB), a blocker of inositol 1,4,5-trisphosphate (IP(3)) receptors and cation channels, and by xestospongin D, a blocker of IP(3) receptors, and was partially blocked by SKF-96365, a non-selective cation channel blocker. However, nicardipine, an L-type voltage-dependent Ca(2+) channel blocker, or N-(p-amylcinnamoyl)anthranilic acid (ACA), a TRPM2 blocker, had little effect on the apoptosis. The inhibitory effect of BAPTA/AM or 2-APB on the H(2)O(2)-induced apoptosis was largely attenuated when the drug was added 30 min or 1 h after start of the treatment with H(2)O(2). These results suggest that the initial intracellular Ca(2+) elevation induced by H(2)O(2), which is mediated via IP(3) receptors and store-operated cation channels, plays an obligatory role in the induction of β-cell apoptosis.
Collapse
Affiliation(s)
- Masahiro Takada
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, University of Shizuoka, 52–1 Yada, Surugaku, Shizuoka, Shizuoka 422–8526, Japan
| | | | | | | | | |
Collapse
|
41
|
Gonzalez A, Santofimia-Castaño P, Rivera-Barreno R, Salido GM. Cinnamtannin B-1, a natural antioxidant that reduces the effects of H(2)O(2) on CCK-8-evoked responses in mouse pancreatic acinar cells. J Physiol Biochem 2011; 68:181-91. [PMID: 22120978 DOI: 10.1007/s13105-011-0130-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 11/07/2011] [Indexed: 01/12/2023]
Abstract
This work was designed in order to gain an insight on the mechanisms by which antioxidants prevent pancreatic disorders. We have examined the properties of cinnamtannin B-1, which belongs to the class of polyphenols, against the effect of hydrogen peroxide (H(2)O(2)) in mouse pancreatic acinar cells. We have studied Ca(2+) mobilization, oxidative state, amylase secretion, and cell viability of cells treated with cinnamtannin B-1 in the presence of various concentrations of H(2)O(2). We found that H(2)O(2) (0.1-100 μM) increased CM-H(2)DCFDA-derived fluorescence, reflecting an increase in oxidation. Cinnamtannin B-1 (10 μM) reduced H(2)O(2)-induced oxidation of CM-H(2)DCFDA. CCK-8 induced oxidation of CM-H(2)DCFDA in a similar way to low micromolar concentrations of H(2)O(2), and cinnamtannin B-1 reduced the oxidant effect of CCK-8. In addition, H(2)O(2) induced a slow and progressive increase in intracellular free Ca(2+) concentration ([Ca(2+)](c)). Cinnamtannin B-1 reduced the effect of H(2)O(2) on [Ca(2+)](c), but only at the lower concentrations of the oxidant. H(2)O(2) inhibited amylase secretion in response to cholecystokinin, and cinnamtannin B-1 reduced the inhibitory action of H(2)O(2) on enzyme secretion. Finally, H(2)O(2) reduced cell viability, and the antioxidant protected acinar cells against H(2)O(2). In conclusion, the beneficial effects of cinnamtannin B-1 appear to be mediated by reducing the intracellular Ca(2+) overload and intracellular accumulation of digestive enzymes evoked by ROS, which is a common pathological precursor that mediates pancreatitis. Our results support the beneficial effect of natural antioxidants in the therapy against oxidative stress-derived deleterious effects on cellular physiology.
Collapse
Affiliation(s)
- Antonio Gonzalez
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, Caceres, Spain.
| | | | | | | |
Collapse
|
42
|
Nazıroğlu M, Çelik Ö, Özgül C, Çiğ B, Doğan S, Bal R, Gümral N, Rodríguez AB, Pariente JA. Melatonin modulates wireless (2.45 GHz)-induced oxidative injury through TRPM2 and voltage gated Ca(2+) channels in brain and dorsal root ganglion in rat. Physiol Behav 2011; 105:683-92. [PMID: 22019785 DOI: 10.1016/j.physbeh.2011.10.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 10/04/2011] [Accepted: 10/05/2011] [Indexed: 01/12/2023]
Abstract
We aimed to investigate the protective effects of melatonin and 2.45 GHz electromagnetic radiation (EMR) on brain and dorsal root ganglion (DRG) neuron antioxidant redox system, Ca(2+) influx, cell viability and electroencephalography (EEG) records in the rat. Thirty two rats were equally divided into four different groups namely group A1: Cage control, group A2: Sham control, group B: 2.45 GHz EMR, group C: 2.45 GHz EMR+melatonin. Groups B and C were exposed to 2.45 GHz EMR during 60 min/day for 30 days. End of the experiments, EEG records and the brain cortex and DRG samples were taken. Lipid peroxidation (LP), cell viability and cytosolic Ca(2+) values in DRG neurons were higher in group B than in groups A1 and A2 although their concentrations were increased by melatonin, 2-aminoethyldiphenyl borinate (2-APB), diltiazem and verapamil supplementation. Spike numbers of EEG records in group C were lower than in group B. Brain cortex vitamin E concentration was higher in group C than in group B. In conclusion, Melatonin supplementation in DRG neurons and brain seems to have protective effects on the 2.45 GHz-induced increase Ca(2+) influx, EEG records and cell viability of the hormone through TRPM2 and voltage gated Ca(2+) channels.
Collapse
Affiliation(s)
- M Nazıroğlu
- Department of Biophysics, Medicine Faculty, Süleyman Demirel University, Isparta, Turkey.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
The endothelium is a highly dynamic structure lining the inside of blood vessels that exhibits physical and chemical properties that are critical determinants of overall vascular function. Physically, the endothelium constitutes a semipermeable barrier. Chemically, the endothelium synthesizes numerous factors such as reactive oxygen species (ROS) that can act as autocrine and paracrine signaling molecules. Oxidative stress results when ROS levels increase to levels that cause cellular injury, and, in the endothelium oxidative stress leads to barrier disruption. Endothelial barrier disruption also results from increased cytosolic calcium through store-operated calcium (SOC) entry channels. Although it is known that ROS can interact with and regulate some ion channels, relatively little is known about the interaction of these species with components of endothelial SOC entry channels, the canonical transient receptor potential (TRPC) proteins. Here we review our current understanding of ROS-mediated TRPC channel function and how it affects SOC entry and endothelial barrier disruption.
Collapse
Affiliation(s)
- Donna L Cioffi
- Department of Biochemistry and Molecular Biology, Center for Lung Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA.
| |
Collapse
|
44
|
Takahashi N, Kozai D, Kobayashi R, Ebert M, Mori Y. Roles of TRPM2 in oxidative stress. Cell Calcium 2011; 50:279-87. [DOI: 10.1016/j.ceca.2011.04.006] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 04/19/2011] [Accepted: 04/22/2011] [Indexed: 12/15/2022]
|
45
|
Glutathione Modulates Ca2+ Influx and Oxidative Toxicity Through TRPM2 Channel in Rat Dorsal Root Ganglion Neurons. J Membr Biol 2011; 242:109-18. [DOI: 10.1007/s00232-011-9382-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2011] [Accepted: 06/17/2011] [Indexed: 01/25/2023]
|
46
|
Neuroprotection by lomerizine, a prophylactic drug for migraine, against hydrogen peroxide-induced hippocampal neurotoxicity. Mol Cell Biochem 2011; 358:1-11. [PMID: 21656126 DOI: 10.1007/s11010-011-0913-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2011] [Accepted: 05/28/2011] [Indexed: 10/18/2022]
Abstract
Migraine is one of the risk factor for ischemic stroke. The purpose of this study was to examine the effect of lomerizine, a prophylactic drug for migraine, on H(2)O(2)-induced cell death of hippocampal neurons. Cytosolic Ca(2+) concentration was measured using fura-2 as a Ca(2+) indicator. Cell death was estimated by trypan blue exclusion. In rat-cultured hippocampal neurons, the addition of H(2)O(2) induced biphasic Ca(2+) elevations and cell death. The H(2)O(2)-induced biphasic Ca(2+) elevations and cell death only occurred when extracellular Ca(2+) was present. The biphasic Ca(2+) elevation was mediated by Ca(2+) influx through the plasma membrane, but not Ca(2+) release from the intracellular Ca(2+) store. Both the early and late phases of H(2)O(2)-induced Ca(2+) influx were reduced by either a T- or L-type voltage-dependent Ca(2+) channel (VDCC) blocker, lomerizine. In fact, L-type VDCC (α(1C) subunit) and T-type VDCC (α(1G) subunit) mRNA were expressed in rat hippocampal neurons. Although an L-type VDCC blocker, nifedipine, partly suppressed the late phase of Ca(2+) influx in response to H(2)O(2), a T-type VDCC blocker, mibefradil, reduced both phases of Ca(2+) influx. Moreover, lomerizine and mibefradil strongly reduced H(2)O(2)-induced cell death, and nifedipine weakly reduced it. These findings suggest that the inhibition of H(2)O(2)-induced Ca(2+) influx through T-type VDCC seems to be important in the protective effect of lomerizine against oxidative stress. It is possible that lomerizine may be a useful drug for prophylactic treatment of migraine, because migraine is a risk factor for ischemic stroke.
Collapse
|
47
|
Freinbichler W, Colivicchi MA, Stefanini C, Bianchi L, Ballini C, Misini B, Weinberger P, Linert W, Varešlija D, Tipton KF, Della Corte L. Highly reactive oxygen species: detection, formation, and possible functions. Cell Mol Life Sci 2011; 68:2067-79. [PMID: 21533983 PMCID: PMC11114910 DOI: 10.1007/s00018-011-0682-x] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 02/27/2011] [Accepted: 03/29/2011] [Indexed: 12/16/2022]
Abstract
The so-called reactive oxygen species (ROS) are defined as oxygen-containing species that are more reactive than O(2) itself, which include hydrogen peroxide and superoxide. Although these are quite stable, they may be converted in the presence of transition metal ions, such as Fe(II), to the highly reactive oxygen species (hROS). hROS may exist as free hydroxyl radicals (HO·), as bound ("crypto") radicals or as Fe(IV)-oxo (ferryl) species and the somewhat less reactive, non-radical species, singlet oxygen. This review outlines the processes by which hROS may be formed, their damaging potential, and the evidence that they might have signaling functions. Since our understanding of the formation and actions of hROS depends on reliable procedures for their detection, particular attention is given to procedures for hROS detection and quantitation and their applicability to in vivo studies.
Collapse
Affiliation(s)
- Wolfhardt Freinbichler
- Institute for Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9/163-AC, 1060 Vienna, Austria
| | - Maria A. Colivicchi
- Dipartimento di Farmacologia Preclinica e Clinica M. Aiazzi Mancini, Università degli Studi di Firenze, Viale G. Pieraccini 6, 50139 Florence, Italy
| | - Chiara Stefanini
- Dipartimento di Farmacologia Preclinica e Clinica M. Aiazzi Mancini, Università degli Studi di Firenze, Viale G. Pieraccini 6, 50139 Florence, Italy
| | - Loria Bianchi
- Dipartimento di Farmacologia Preclinica e Clinica M. Aiazzi Mancini, Università degli Studi di Firenze, Viale G. Pieraccini 6, 50139 Florence, Italy
- Present Address: Azienda USL 3 di Pistoia, 51100 Pistoia, Italy
| | - Chiara Ballini
- Dipartimento di Farmacologia Preclinica e Clinica M. Aiazzi Mancini, Università degli Studi di Firenze, Viale G. Pieraccini 6, 50139 Florence, Italy
| | - Bashkim Misini
- Institute for Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9/163-AC, 1060 Vienna, Austria
| | - Peter Weinberger
- Institute for Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9/163-AC, 1060 Vienna, Austria
| | - Wolfgang Linert
- Institute for Applied Synthetic Chemistry, Vienna University of Technology, Getreidemarkt 9/163-AC, 1060 Vienna, Austria
| | - Damir Varešlija
- Department of Biochemistry, Trinity College, Dublin 2, Ireland
| | - Keith F. Tipton
- Department of Biochemistry, Trinity College, Dublin 2, Ireland
| | - Laura Della Corte
- Dipartimento di Farmacologia Preclinica e Clinica M. Aiazzi Mancini, Università degli Studi di Firenze, Viale G. Pieraccini 6, 50139 Florence, Italy
| |
Collapse
|
48
|
Nazıroğlu M. TRPM2 channel membrane currents in primary rat megakaryocytes were activated by the agonist ADP-ribose but not oxidative stress. J Membr Biol 2011; 241:51-7. [PMID: 21512734 DOI: 10.1007/s00232-011-9356-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Accepted: 03/07/2011] [Indexed: 01/14/2023]
Abstract
Melastatin-like transient receptor potential 2 (TRPM2) channel activation/inhibition mechanisms in response to ADP-ribose (ADPR), oxidative stress, flufenamic acid (FFA) and 2-aminoethoxydiphenyl borate (2-APB) are not clear. We tested the effects of FFA and 2-APB on ADPR-induced TRPM2 cation channel currents in rat native bone marrow megakaryocytes. Megakaryocyte cells were freshly isolated from rat bone marrow and studied with the conventional whole-cell patch-clamp technique. Extracellular H2O2, FFA and 2-APB were added through the patch chamber, while intracellular ADPR was applied through the pipette. Nonselective cation currents were consistently induced by ADPR but not H2O2. Current density of ADPR in the cells was significantly (P<0.001) higher than in control. The time courses of ADPR effects in the megakaryocytes were characterized by a delay of 2.24±0.73. The ADPR-induced Ca2+ gate was not blocked by either the IP3 receptor inhibitor 2-APB or the PLC inhibitor FFA. In conclusion, TRPM2 channels were constitutively activated by intracellular ADPR, although the channel currents in rat native megakaryocytes were not affected by extracellular H2O2, 2-APB or FFA. Activation of TRPM2 channels in megakaryocytes seems to be intracellular and ADPR-dependent.
Collapse
Affiliation(s)
- Mustafa Nazıroğlu
- Department of Biophysics, Medical Faculty, Süleyman Demirel University, Morfoloji Binasi, 32260, Isparta, Turkey.
| |
Collapse
|
49
|
Aminoethoxydiphenyl borate and flufenamic acid inhibit Ca2+ influx through TRPM2 channels in rat dorsal root ganglion neurons activated by ADP-ribose and rotenone. J Membr Biol 2011; 241:69-75. [PMID: 21509529 DOI: 10.1007/s00232-011-9363-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2011] [Accepted: 03/21/2011] [Indexed: 12/21/2022]
Abstract
Exposure to oxidative stress causes health problems, including sensory neuron neuropathy and pain. Rotenone is a toxin used to generate intracellular oxidative stress in neurons. However, the mechanism of toxicity in dorsal root ganglion (DRG) neurons has not been characterized. Melastatin-like transient receptor potential 2 (TRPM2) channel activation and inhibition in response to oxidative stress, ADP-ribose (ADPR), flufenamic acid (FFA) and 2-aminoethoxydiphenyl borate (2-APB) in DRG neurons are also not clear. We tested the effects of FFA and 2-APB on ADPR and rotenone-induced TRPM2 cation channel activation in DRG neurons of rats. DRG neurons were freshly isolated from rats and studied with the conventional whole-cell patch-clamp technique. Rotenone, FFA and 2-APB were extracellularly added through the patch chamber, and ADPR was applied intracellularly through the patch pipette. TRPM2 cation currents were consistently induced by ADPR and rotenone. Current densities of the neurons were higher in the ADPR and rotenone groups than in control. The time courses (gating times) in the neurons were longer in the rotenone than in the ADPR group. ADPR and rotenone-induced TRPM2 currents were totally blocked by 2-APB and partially blocked by FFA. In conclusion, TRPM2 channels were constitutively activated by ADPR and rotenone, and 2-APB and FFA induced an inhibitory effect on TRPM2 cation channel currents in rat DRG neurons. Since oxidative stress is a common feature of neuropathic pain and diseases of sensory neurons, the present findings have broad application to the etiology of neuropathic pain and diseases of DRG neurons.
Collapse
|
50
|
Grape Seed Extract Alleviates High-Fat Diet-Induced Obesity and Heart Dysfunction by Preventing Cardiac Siderosis. Cardiovasc Toxicol 2011; 11:28-37. [DOI: 10.1007/s12012-010-9101-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|