1
|
Stein CS, Linzer CR, Heer CD, Witmer NH, Cochran JD, Spitz DR, Boudreau RL. Mitoregulin Promotes Cell Cycle Progression in Non-Small Cell Lung Cancer Cells. Int J Mol Sci 2025; 26:1939. [PMID: 40076565 PMCID: PMC11899852 DOI: 10.3390/ijms26051939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Mitoregulin (MTLN) is a 56-amino-acid mitochondrial microprotein known to modulate mitochondrial energetics. MTLN gene expression is elevated broadly across most cancers and has been proposed as a prognostic biomarker for non-small cell lung cancer (NSCLC). In addition, lower MTLN expression in lung adenocarcinoma (LUAD) correlates with significantly improved patient survival. In our studies, we have found that MTLN silencing in A549 NSCLC cells slowed proliferation and, in accordance with this, we observed the following: (1) increased proportion of cells in the G1 phase of cell cycle; (2) protein changes consistent with G1 arrest (e.g., reduced levels and/or reduced phosphorylation of ERK, MYC, CDK2, and RB, and elevated p27Kip1); (3) reduction in clonogenic cell survival and; (4) lower steady-state cytosolic and mitochondrial H2O2 levels as indicated by use of the roGFP2-Orp1 redox sensor. Conflicting with G1 arrest, we observed a boost in cyclin D1 abundance. We also tested MTLN silencing in combination with buthionine sulfoximine (BSO) and auranofin (AF), drugs that inhibit GSH synthesis and thioredoxin reductase, respectively, to elevate the reactive oxygen species (ROS) amount to a toxic range. Interestingly, clonogenic survival after drug treatment was greater for MTLN-silenced cultures versus the control cultures. Lower H2O2 output and reduced vulnerability to ROS damage due to G1 status may have jointly contributed to the partial BSO + AF resistance. Overall, our results provide evidence that MTLN fosters H2O2 signaling to propel G1/S transition and suggest MTLN silencing as a therapeutic strategy to limit NSCLC growth.
Collapse
Affiliation(s)
- Colleen S. Stein
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (C.R.L.); (N.H.W.); (J.D.C.)
| | - Connor R. Linzer
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (C.R.L.); (N.H.W.); (J.D.C.)
| | - Collin D. Heer
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; (C.D.H.); (D.R.S.)
| | - Nathan H. Witmer
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (C.R.L.); (N.H.W.); (J.D.C.)
| | - Jesse D. Cochran
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (C.R.L.); (N.H.W.); (J.D.C.)
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; (C.D.H.); (D.R.S.)
| | - Ryan L. Boudreau
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (C.R.L.); (N.H.W.); (J.D.C.)
| |
Collapse
|
2
|
Chen HR, Sun Y, Mittler G, Rumpf T, Shvedunova M, Grosschedl R, Akhtar A. MOF-mediated PRDX1 acetylation regulates inflammatory macrophage activation. Cell Rep 2024; 43:114682. [PMID: 39207899 DOI: 10.1016/j.celrep.2024.114682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 06/27/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
Signaling-dependent changes in protein phosphorylation are critical to enable coordination of transcription and metabolism during macrophage activation. However, the role of acetylation in signal transduction during macrophage activation remains obscure. Here, we identify the redox signaling regulator peroxiredoxin 1 (PRDX1) as a substrate of the lysine acetyltransferase MOF. MOF acetylates PRDX1 at lysine 197, preventing hyperoxidation and thus maintaining its activity under stress. PRDX1 K197ac responds to inflammatory signals, decreasing rapidly in mouse macrophages stimulated with bacterial lipopolysaccharides (LPSs) but not with interleukin (IL)-4 or IL-10. The LPS-induced decrease of PRDX1 K197ac elevates cellular hydrogen peroxide accumulation and augments ERK1/2, but not p38 or AKT, phosphorylation. Concomitantly, diminished PRDX1 K197ac stimulates glycolysis, potentiates H3 serine 28 phosphorylation, and ultimately enhances the production of pro-inflammatory mediators such as IL-6. Our work reveals a regulatory role for redox protein acetylation in signal transduction and coordinating metabolic and transcriptional programs during inflammatory macrophage activation.
Collapse
Affiliation(s)
- Hui-Ru Chen
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany; Albert-Ludwigs-University Freiburg, Faculty of Biology, Freiburg, Baden-Württemberg, Germany
| | - Yidan Sun
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany
| | - Gerhard Mittler
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany
| | - Tobias Rumpf
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany
| | - Maria Shvedunova
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany
| | - Rudolf Grosschedl
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany
| | - Asifa Akhtar
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Württemberg, Germany.
| |
Collapse
|
3
|
Wei S, Ju F, Xiao J, Li J, Liu T, Hu Z. Aloperine Alleviates Myocardial Injury Induced by Myocardial Ischemia and Reperfusion by Activating the ERK1/2/β-catenin Signaling Pathway. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07566-0. [PMID: 38416285 DOI: 10.1007/s10557-024-07566-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/21/2024] [Indexed: 02/29/2024]
Abstract
OBJECTIVE Myocardial ischemia/reperfusion (I/R) injury can cause severe cardiac damage. Aloperine is a quinolizidine alkaloid found in the leaves and seeds of Sophora alopecuroides L. It has been recognized that aloperine has organ-protective properties; however, its role in cardioprotection is poorly characterized. This study aimed to evaluate the cardioprotective effects of aloperine against myocardial I/R injury in vivo. METHODS Adult male Sprague‒Dawley rats were randomly divided into sham-operated, control, and aloperine groups. All rats except for the sham-operated rats were subjected to 45 min of myocardial ischemia (by left anterior descending ligation) followed by 3 h of reperfusion. Aloperine (10 mg/kg) was given intravenously at the onset of reperfusion. The cardioprotective effects of aloperine were evaluated by determining infarct size, hemodynamics, histological changes, cardiac biomarkers, and cardiac apoptosis. RESULTS Aloperine limited infarct size; improved hemodynamics; attenuated myocardial I/R-induced histological deterioration; decreased serum LDH, CK-MB, and α-HBDH levels; and inhibited apoptosis after myocardial I/R injury. Moreover, aloperine stimulated the phosphorylation of ventricular ERK1/2, which is a major module of MAPK signaling pathways. Furthermore, aloperine increased the ventricular expression levels of β-catenin. Pharmacological inhibition of ERK1/2 diminished aloperine-induced cardioprotection and blocked ERK1/2/β-catenin signaling. CONCLUSIONS These data support the cardioprotective effect of aloperine against myocardial I/R injury, which is mediated, at least in part, by the ERK1/2/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Shichao Wei
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Ju
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junshen Xiao
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiaxue Li
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ting Liu
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhaoyang Hu
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
4
|
Schaffer AM, Fiala GJ, Hils M, Natali E, Babrak L, Herr LA, Romero-Mulero MC, Cabezas-Wallscheid N, Rizzi M, Miho E, Schamel WWA, Minguet S. Kidins220 regulates the development of B cells bearing the λ light chain. eLife 2024; 13:e83943. [PMID: 38271217 PMCID: PMC10810608 DOI: 10.7554/elife.83943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/27/2023] [Indexed: 01/27/2024] Open
Abstract
The ratio between κ and λ light chain (LC)-expressing B cells varies considerably between species. We recently identified Kinase D-interacting substrate of 220 kDa (Kidins220) as an interaction partner of the BCR. In vivo ablation of Kidins220 in B cells resulted in a marked reduction of λLC-expressing B cells. Kidins220 knockout B cells fail to open and recombine the genes of the Igl locus, even in genetic scenarios where the Igk genes cannot be rearranged or where the κLC confers autoreactivity. Igk gene recombination and expression in Kidins220-deficient B cells is normal. Kidins220 regulates the development of λLC B cells by enhancing the survival of developing B cells and thereby extending the time-window in which the Igl locus opens and the genes are rearranged and transcribed. Further, our data suggest that Kidins220 guarantees optimal pre-BCR and BCR signaling to induce Igl locus opening and gene recombination during B cell development and receptor editing.
Collapse
Affiliation(s)
- Anna-Maria Schaffer
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Signalling Research Centers BIOSS and CIBSS, University of FreiburgFreiburgGermany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
| | - Gina Jasmin Fiala
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Signalling Research Centers BIOSS and CIBSS, University of FreiburgFreiburgGermany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
| | - Miriam Hils
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of MunichMunichGermany
| | - Eriberto Natali
- Institute of Medical Engineering and Medical Informatics, School of Life Sciences, FHNW 15 University of Applied Sciences and Arts Northwestern SwitzerlandMuttenzSwitzerland
| | - Lmar Babrak
- Institute of Medical Engineering and Medical Informatics, School of Life Sciences, FHNW 15 University of Applied Sciences and Arts Northwestern SwitzerlandMuttenzSwitzerland
| | - Laurenz Alexander Herr
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Signalling Research Centers BIOSS and CIBSS, University of FreiburgFreiburgGermany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
| | - Mari Carmen Romero-Mulero
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Nina Cabezas-Wallscheid
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
- CIBSS – Centre for Integrative Biological Signalling Studies, University of FreiburgFreiburgGermany
| | - Marta Rizzi
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
- CIBSS – Centre for Integrative Biological Signalling Studies, University of FreiburgFreiburgGermany
- Division of Clinical and Experimental Immunology, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of ViennaViennaAustria
- Department of Rheumatology and Clinical Immunology, University Medical Center Freiburg, Faculty of Medicine, University of FreiburgFreiburgGermany
| | - Enkelejda Miho
- Institute of Medical Engineering and Medical Informatics, School of Life Sciences, FHNW 15 University of Applied Sciences and Arts Northwestern SwitzerlandMuttenzSwitzerland
- aiNET GmbHBaselSwitzerland
- SIB Swiss Institute of BioinformaticsLausanneSwitzerland
| | - Wolfgang WA Schamel
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Signalling Research Centers BIOSS and CIBSS, University of FreiburgFreiburgGermany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
| | - Susana Minguet
- Faculty of Biology, Albert-Ludwigs-University of FreiburgFreiburgGermany
- Signalling Research Centers BIOSS and CIBSS, University of FreiburgFreiburgGermany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical FacultyFreiburgGermany
| |
Collapse
|
5
|
Behera R, Sharma V, Grewal AK, Kumar A, Arora B, Najda A, Albadrani GM, Altyar AE, Abdel-Daim MM, Singh TG. Mechanistic correlation between mitochondrial permeability transition pores and mitochondrial ATP dependent potassium channels in ischemia reperfusion. Biomed Pharmacother 2023; 162:114599. [PMID: 37004326 DOI: 10.1016/j.biopha.2023.114599] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023] Open
Abstract
Mitochondrial dysfunction is one of the fundamental causes of ischemia reperfusion (I/R) damage. I/R refers to the paradoxical progression of cellular dysfunction and death that occurs when blood flow is restored to previously ischemic tissues. I/R causes a significant rise in mitochondrial permeability resulting in the opening of mitochondrial permeability transition pores (MPTP). The MPTP are broad, nonspecific channels present in the inner mitochondrial membrane (IMM), and are known to mediate the deadly permeability alterations that trigger mitochondrial driven cell death. Protection from reperfusion injury occurs when long-term ischemia is accompanied by short-term ischemic episodes or inhibition of MPTP from opening via mitochondrial ATP dependent potassium (mitoKATP) channels. These channels located in the IMM, play an essential role in ischemia preconditioning (PC) and protect against cell death by blocking MPTP opening. This review primarily focuses on the interaction between the MPTP and mitoKATP along with their role in the I/R injury. This article also describes the molecular composition of the MPTP and mitoKATP in order to promote future knowledge and treatment of diverse I/R injuries in various organs.
Collapse
|
6
|
Modesti L, Danese A, Angela Maria Vitto V, Ramaccini D, Aguiari G, Gafà R, Lanza G, Giorgi C, Pinton P. Mitochondrial Ca 2+ Signaling in Health, Disease and Therapy. Cells 2021; 10:cells10061317. [PMID: 34070562 PMCID: PMC8230075 DOI: 10.3390/cells10061317] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 12/12/2022] Open
Abstract
The divalent cation calcium (Ca2+) is considered one of the main second messengers inside cells and acts as the most prominent signal in a plethora of biological processes. Its homeostasis is guaranteed by an intricate and complex system of channels, pumps, and exchangers. In this context, by regulating cellular Ca2+ levels, mitochondria control both the uptake and release of Ca2+. Therefore, at the mitochondrial level, Ca2+ plays a dual role, participating in both vital physiological processes (ATP production and regulation of mitochondrial metabolism) and pathophysiological processes (cell death, cancer progression and metastasis). Hence, it is not surprising that alterations in mitochondrial Ca2+ (mCa2+) pathways or mutations in Ca2+ transporters affect the activities and functions of the entire cell. Indeed, it is widely recognized that dysregulation of mCa2+ signaling leads to various pathological scenarios, including cancer, neurological defects and cardiovascular diseases (CVDs). This review summarizes the current knowledge on the regulation of mCa2+ homeostasis, the related mechanisms and the significance of this regulation in physiology and human diseases. We also highlight strategies aimed at remedying mCa2+ dysregulation as promising therapeutical approaches.
Collapse
Affiliation(s)
- Lorenzo Modesti
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Alberto Danese
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Veronica Angela Maria Vitto
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Daniela Ramaccini
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Gianluca Aguiari
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy;
| | - Roberta Gafà
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (R.G.); (G.L.)
| | - Giovanni Lanza
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (R.G.); (G.L.)
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
| | - Paolo Pinton
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (L.M.); (A.D.); (V.A.M.V.); (D.R.); (C.G.)
- Correspondence: ; Tel.: +39-0532-455802
| |
Collapse
|
7
|
Xue S, Tang H, Zhao G, Fang C, Shen Y, Yan D, Yuan Y, Fu W, Shi Z, Tang X, Guo D. C-C motif ligand 8 promotes atherosclerosis via NADPH oxidase 2/reactive oxygen species-induced endothelial permeability increase. Free Radic Biol Med 2021; 167:181-192. [PMID: 33741452 DOI: 10.1016/j.freeradbiomed.2021.02.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/29/2021] [Accepted: 02/14/2021] [Indexed: 12/31/2022]
Abstract
Chemokines have been reported to play important roles in atherosclerotic development. Recently, we found C-C motif ligand 8 (CCL8), a rarely studied chemokine in atherosclerosis, was highly expressed in the endothelium of advanced human carotid plaques. We hypothesized whether CCL8 promotes atherosclerosis through endothelial dysfunction. Apolipoprotein E-deficient mice under the Western diet were used to construct atherosclerosis models. Adeno-associated viruses (AAV) with CCL8 and the CCL8-antibody were injected into mice respectively to conduct CCL8 overexpression and suppression. The results showed that atherosclerotic lesions were significantly increased in the AAV-CCL8 group, while, lesions in the aortic sinus were reduced in the CCL8-antibody group. With CCL8 treatment (200 ng/ml, 24 h) in vitro, the permeability of human aortic endothelial cells (HAECs) increased and the expression of junctional proteins Zonula occluden-1, and Vascular endothelial cadherin were decreased. This effect was dependent on reactive oxygen species (ROS) generation, which could be blocked by l-Ascorbic acid and Apocynin. Results showed that NADPH oxidase 2 (NOX2) expression also increased with CCL8 stimulation and the ROS, and permeability increase of HAECs could be inhibited when NOX2 interfered with the specific siRNA. Additionally, we further found ERK1/2, PI3K-AKT, and NF-κB pathways were involved in the activation of CCL8. Our results indicated that CCL8 might also play important roles in atherosclerosis and this effect, at least in part, was caused by NOX2/ROS-induced endothelial permeability increase. This study might contribute to a deeper understanding of the connection between chemokines and atherosclerosis.
Collapse
Affiliation(s)
- Song Xue
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hanfei Tang
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gefei Zhao
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Jiansu, China
| | - Chao Fang
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yang Shen
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dong Yan
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ye Yuan
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weiguo Fu
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhenyu Shi
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao Tang
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Daqiao Guo
- Department of Vascular Surgery, Institute of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
8
|
Nakamura-Ishizu A, Ito K, Suda T. Hematopoietic Stem Cell Metabolism during Development and Aging. Dev Cell 2021; 54:239-255. [PMID: 32693057 DOI: 10.1016/j.devcel.2020.06.029] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/02/2020] [Accepted: 06/26/2020] [Indexed: 12/22/2022]
Abstract
Cellular metabolism in hematopoietic stem cells (HSCs) is an area of intense research interest, but the metabolic requirements of HSCs and their adaptations to their niches during development have remained largely unaddressed. Distinctive from other tissue stem cells, HSCs transition through multiple hematopoietic sites during development. This transition requires drastic metabolic shifts, insinuating the capacity of HSCs to meet the physiological demand of hematopoiesis. In this review, we highlight how mitochondrial metabolism determines HSC fate, and especially focus on the links between mitochondria, endoplasmic reticulum (ER), and lysosomes in HSC metabolism.
Collapse
Affiliation(s)
- Ayako Nakamura-Ishizu
- Department of Microscopic and Developmental Anatomy, Tokyo Women's Medical University, 8-1 Kawadacho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Keisuke Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, 1301 Morris Park Ave., Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA; Department of Medicine (Hemato-Oncology), Montefiore Medical Center, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA; Albert Einstein Cancer Center and Diabetes Research Center, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY, USA
| | - Toshio Suda
- Cancer Science Institute, National University of Singapore, 14 Medical Drive, MD6, 117599 Singapore, Singapore; International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto City 860-0811, Japan.
| |
Collapse
|
9
|
Abstract
Significance: Cancer cells are stabilized in an undifferentiated state similar to stem cells. This leads to profound modifications of their metabolism, which further modifies their genetics and epigenetics as malignancy progresses. Specific metabolites and enzymes may serve as clinical markers of cancer progression. Recent Advances: Both 2-hydroxyglutarate (2HG) enantiomers are associated with reprogrammed metabolism, in grade III/IV glioma, glioblastoma, and acute myeloid leukemia cells, and numerous other cancer types, while acting also in the cross talk of tumors with immune cells. 2HG contributes to specific alternations in cancer metabolism and developed oxidative stress, while also inducing decisions on the differentiation of naive T lymphocytes, and serves as a signal messenger in immune cells. Moreover, 2HG inhibits chromatin-modifying enzymes, namely 2-oxoglutarate-dependent dioxygenases, and interferes with hypoxia-inducible factor (HIF) transcriptome reprogramming and mammalian target of rapamycin (mTOR) pathway, thus dysregulating gene expression and further promoting cancerogenesis. Critical Issues: Typically, heterozygous mutations within the active sites of isocitrate dehydrogenase isoform 1 (IDH1)R132H and mitochondrial isocitrate dehydrogenase isoform 2 (IDH2)R140Q provide cells with millimolar r-2-hydroxyglutarate (r-2HG) concentrations, whereas side activities of lactate and malate dehydrogenase form submillimolar s-2-hydroxyglutarate (s-2HG). However, even wild-type IDH1 and IDH2, notably under shifts toward reductive carboxylation glutaminolysis or changes in other enzymes, lead to "intermediate" 0.01-0.1 mM 2HG levels, for example, in breast carcinoma compared with 10-8M in noncancer cells. Future Directions: Uncovering further molecular metabolism details specific for given cancer cell types and sequence-specific epigenetic alternations will lead to the design of diagnostic approaches, not only for predicting patients' prognosis or uncovering metastases and tumor remissions but also for early diagnostics.
Collapse
Affiliation(s)
- Petr Ježek
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
10
|
Ohwada W, Tanno M, Yano T, Ong SB, Abe K, Sato T, Kuno A, Miki T, Sugawara H, Igaki Y, Miura T. Distinct intra-mitochondrial localizations of pro-survival kinases and regulation of their functions by DUSP5 and PHLPP-1. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165851. [PMID: 32480039 DOI: 10.1016/j.bbadis.2020.165851] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/14/2020] [Accepted: 05/26/2020] [Indexed: 01/03/2023]
Abstract
ERK and Akt have been shown to regulate cell sensitivity to death-inducing stress by phosphorylating GSK-3β, a major modulator of the threshold for mitochondrial permeability transition. Here we examined intra-mitochondrial localization of the pro-survival kinases and their regulation by phosphatases. Stepwise trypsin digestion of mitochondria isolated from HEK293 or H9c2 cells was performed, and immunoblotting revealed that GSK-3β and ERK localized dominantly in the outer membrane (OM), while Akt resided at comparable levels in OM, the inner membrane (IM) and the matrix. Treatment with IGF-1 increased the protein level of Akt in the matrix, while ERK and GSK-3β protein levels were increased in OM. Simultaneously, IGF-1 treatment elevated the level of Thr202/Tyr204-phospho-ERK in IM and matrix and levels of Ser473-phospho-Akt and Ser9-phospho-GSK-3β in OM, IM and matrix. Exposing cells to reactive oxygen species (ROS) by using antimycin A increased the levels of DUSP5 and PHLPP-1 mainly in OM and induced dephosphorylation of Akt, ERK and GSK-3β. The mitochondrial localization of DUSP5 was confirmed by experiments with mitochondria purified by Percoll gradient centrifugation and by transfection of cells with GFP-tagged DUSP5. Knockdown of either DUSP5 or PHLPP-1 increased the levels of both Thr202/Tyr204-phospho-ERK and Ser473-phospho-Akt in mitochondria. Cell death induced by antimycin A was suppressed by siRNA-mediated knockdown of DUSP5. The results suggest that Akt and ERK in mitochondria show distinct intra-mitochondrial localization and crosstalk in GSK-3β regulation and that recruitment of DUSP5 as well as PHLPP-1 to mitochondria contributes to ROS-induced termination of the protective signaling.
Collapse
Affiliation(s)
- Wataru Ohwada
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masaya Tanno
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshiyuki Yano
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Sang-Bing Ong
- Signature Research Program in Cardiovascular & Metabolic Diseases, Duke-NUS Medical School, Singapore
| | - Koki Abe
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tatsuya Sato
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan; Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Atsushi Kuno
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan; Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takayuki Miki
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hirohito Sugawara
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yusuke Igaki
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tetsuji Miura
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.
| |
Collapse
|
11
|
Margaritelis NV, Paschalis V, Theodorou AA, Kyparos A, Nikolaidis MG. Redox basis of exercise physiology. Redox Biol 2020; 35:101499. [PMID: 32192916 PMCID: PMC7284946 DOI: 10.1016/j.redox.2020.101499] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/20/2020] [Accepted: 03/05/2020] [Indexed: 12/15/2022] Open
Abstract
Redox reactions control fundamental processes of human biology. Therefore, it is safe to assume that the responses and adaptations to exercise are, at least in part, mediated by redox reactions. In this review, we are trying to show that redox reactions are the basis of exercise physiology by outlining the redox signaling pathways that regulate four characteristic acute exercise-induced responses (muscle contractile function, glucose uptake, blood flow and bioenergetics) and four chronic exercise-induced adaptations (mitochondrial biogenesis, muscle hypertrophy, angiogenesis and redox homeostasis). Based on our analysis, we argue that redox regulation should be acknowledged as central to exercise physiology.
Collapse
Affiliation(s)
- N V Margaritelis
- Department of Physical Education and Sport Science at Serres, Aristotle University of Thessaloniki, Thessaloniki, Greece; Dialysis Unit, 424 General Military Hospital of Thessaloniki, Thessaloniki, Greece.
| | - V Paschalis
- School of Physical Education and Sport Science, National and Kapodistrian University of Athens, Athens, Greece
| | - A A Theodorou
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - A Kyparos
- Department of Physical Education and Sport Science at Serres, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - M G Nikolaidis
- Department of Physical Education and Sport Science at Serres, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| |
Collapse
|
12
|
Zhang LM, Zhang DX, Zhao XC, Sun W. RETRACTED ARTICLE: Erythropoietin Rescues Primary Rat Cortical Neurons by Altering the Nrf2:Bach1 Ratio: Roles of Extracellular Signal-Regulated Kinase 1/2. Neurochem Res 2020; 45:1244. [PMID: 28083849 DOI: 10.1007/s11064-017-2174-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 12/31/2016] [Accepted: 01/03/2017] [Indexed: 01/01/2023]
Affiliation(s)
- Li-Min Zhang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China.
| | - Dong-Xue Zhang
- Department of Gerontology, Cangzhou Central Hospital, Cangzhou, China
| | - Xiao-Chun Zhao
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Wenbo Sun
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, China
| |
Collapse
|
13
|
Aluksanasuwan S, Plumworasawat S, Malaitad T, Chaiyarit S, Thongboonkerd V. High glucose induces phosphorylation and oxidation of mitochondrial proteins in renal tubular cells: A proteomics approach. Sci Rep 2020; 10:5843. [PMID: 32246012 PMCID: PMC7125224 DOI: 10.1038/s41598-020-62665-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 03/18/2020] [Indexed: 12/11/2022] Open
Abstract
Mitochondrial dysfunction has been thought to play roles in the pathogenesis of diabetic nephropathy (DN). However, precise mechanisms underlying mitochondrial dysfunction in DN remained unclear. Herein, mitochondria were isolated from renal tubular cells after exposure to normal glucose (5.5 mM glucose), high glucose (25 mM glucose), or osmotic control (5.5 mM glucose + 19.5 mM mannitol) for 96 h. Comparative proteomic analysis revealed six differentially expressed proteins among groups that were subsequently identified by tandem mass spectrometry (nanoLC-ESI-ETD MS/MS) and confirmed by Western blotting. Several various types of post-translational modifications (PTMs) were identified in all of these identified proteins. Interestingly, phosphorylation and oxidation were most abundant in mitochondrial proteins whose levels were exclusively increased in high glucose condition. The high glucose-induced increases in phosphorylation and oxidation of mitochondrial proteins were successfully confirmed by various assays including MS/MS analyses. Moreover, high glucose also increased levels of phosphorylated ezrin, intracellular ATP and ROS, all of which could be abolished by a p38 MAPK inhibitor (SB239063), implicating a role of p38 MAPK-mediated phosphorylation in high glucose-induced mitochondrial dysfunction. These data indicate that phosphorylation and oxidation of mitochondrial proteins are, at least in part, involved in mitochondrial dysfunction in renal tubular cells during DN.
Collapse
Affiliation(s)
- Siripat Aluksanasuwan
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Sirikanya Plumworasawat
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Thanyalak Malaitad
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Sakdithep Chaiyarit
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
14
|
Helfenberger KE, Castillo AF, Mele PG, Fiore A, Herrera L, Finocchietto P, Podestá EJ, Poderoso C. Angiotensin II stimulation promotes mitochondrial fusion as a novel mechanism involved in protein kinase compartmentalization and cholesterol transport in human adrenocortical cells. J Steroid Biochem Mol Biol 2019; 192:105413. [PMID: 31202858 DOI: 10.1016/j.jsbmb.2019.105413] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 05/10/2019] [Accepted: 06/13/2019] [Indexed: 01/22/2023]
Abstract
In steroid-producing cells, cholesterol transport from the outer to the inner mitochondrial membrane is the first and rate-limiting step for the synthesis of all steroid hormones. Cholesterol can be transported into mitochondria by specific mitochondrial protein carriers like the steroidogenic acute regulatory protein (StAR). StAR is phosphorylated by mitochondrial ERK in a cAMP-dependent transduction pathway to achieve maximal steroid production. Mitochondria are highly dynamic organelles that undergo replication, mitophagy and morphology changes, all processes allowed by mitochondrial fusion and fission, known as mitochondrial dynamics. Mitofusin (Mfn) 1 and 2 are GTPases involved in the regulation of fusion, while dynamin-related protein 1 (Drp1) is the major regulator of mitochondrial fission. Despite the role of mitochondrial dynamics in neurological and endocrine disorders, little is known about fusion/fission in steroidogenic tissues. In this context, the present work aimed to study the role of angiotensin II (Ang II) in protein subcellular compartmentalization, mitochondrial dynamics and the involvement of this process in the regulation of aldosterone synthesis. We demonstrate here that Ang II stimulation promoted the recruitment and activation of PKCε, ERK and its upstream kinase MEK to the mitochondria, all of them essential for steroid synthesis. Moreover, Ang II prompted a shift from punctate to tubular/elongated (fusion) mitochondrial shape, in line with the observation of hormone-dependent upregulation of Mfn2 levels. Concomitantly, mitochondrial Drp1 was diminished, driving mitochondria toward fusion. Moreover, Mfn2 expression is required for StAR, ERK and MEK mitochondrial localization and ultimately for aldosterone synthesis. Collectively, this study provides fresh insights into the importance of hormonal regulation in mitochondrial dynamics as a novel mechanism involved in aldosterone production.
Collapse
Affiliation(s)
- Katia E Helfenberger
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Paraguay 2155 5th floor, C1121ABG, Ciudad de Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Ciudad de Buenos Aires, Argentina
| | - Ana F Castillo
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Paraguay 2155 5th floor, C1121ABG, Ciudad de Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Ciudad de Buenos Aires, Argentina
| | - Pablo G Mele
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Paraguay 2155 5th floor, C1121ABG, Ciudad de Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Ciudad de Buenos Aires, Argentina
| | - Ana Fiore
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Paraguay 2155 5th floor, C1121ABG, Ciudad de Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Ciudad de Buenos Aires, Argentina
| | - Lucía Herrera
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Paraguay 2155 5th floor, C1121ABG, Ciudad de Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Ciudad de Buenos Aires, Argentina
| | - Paola Finocchietto
- Universidad de Buenos Aires, Facultad de Medicina, Hospital de Clínicas "José de San Martín", Laboratorio del Metabolismo del Oxígeno, Av. Córdoba 2351, C1121ABJ, Ciudad de Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo (INIGEM), Ciudad de Buenos Aires, Argentina
| | - Ernesto J Podestá
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Paraguay 2155 5th floor, C1121ABG, Ciudad de Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Ciudad de Buenos Aires, Argentina
| | - Cecilia Poderoso
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Paraguay 2155 5th floor, C1121ABG, Ciudad de Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Ciudad de Buenos Aires, Argentina.
| |
Collapse
|
15
|
Chen PJ, Ko IL, Lee CL, Hu HC, Chang FR, Wu YC, Leu YL, Wu CC, Lin CY, Pan CY, Tsai YF, Hwang TL. Targeting allosteric site of AKT by 5,7-dimethoxy-1,4-phenanthrenequinone suppresses neutrophilic inflammation. EBioMedicine 2019; 40:528-540. [PMID: 30709770 PMCID: PMC6413683 DOI: 10.1016/j.ebiom.2019.01.043] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/22/2019] [Accepted: 01/22/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Acute lung injury (ALI) is a severe life-threatening inflammatory disease. Neutrophil activation is a major pathogenic factor in ALI. Protein kinase B (PKB)/AKT regulates diverse cellular responses, but the significance in neutrophilic inflammation and ALI remains unknown. METHODS Human neutrophils and neutrophil-like differentiated HL-60 (dHL-60) cells were used to examine the anti-inflammatory effects of 5,7-dimethoxy-1,4-phenanthrenequinone (CLLV-1). The therapeutic potential of CLLV-1 was determined in a mouse model of lipopolysaccharide (LPS)-induced ALI. FINDINGS CLLV-1 inhibited respiratory burst, degranulation, adhesion, and chemotaxis in human neutrophils and dHL-60 cells. CLLV-1 inhibited the phosphorylation of AKT (Thr308 and Ser473), but not of ERK, JNK, or p38. Furthermore, CLLV-1 blocked AKT activity and covalently reacted with AKT Cys310 in vitro. The AKT309-313 peptide-CLLV-1 adducts were determined by NMR or mass spectrometry assay. The alkylation agent-conjugated AKT (reduced form) level was also inhibited by CLLV-1. Significantly, CLLV-1 ameliorated LPS-induced ALI, neutrophil infiltration, and AKT activation in mice. INTERPRETATION Our results identify CLLV-1 as a covalent allosteric AKT inhibitor by targeting AKT Cys310. CLLV-1 shows potent anti-inflammatory activity in human neutrophils and LPS-induced mouse ALI. Our findings provide a mechanistic framework for redox modification of AKT that may serve as a novel pharmacological target to alleviate neutrophilic inflammation.
Collapse
Affiliation(s)
- Po-Jen Chen
- Department of Cosmetic Science, Providence University, Taichung 433, Taiwan; Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - I-Ling Ko
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chia-Lin Lee
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung 404, Taiwan; Department of Cosmeceutics, China Medical University, Taichung 404, Taiwan
| | - Hao-Chun Hu
- Graduate Institute of Natural Products, College of Pharmacy and Research Center for Natural Products & Drug Development, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, College of Pharmacy and Research Center for Natural Products & Drug Development, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yang-Chang Wu
- Graduate Institute of Natural Products, College of Pharmacy and Research Center for Natural Products & Drug Development, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yann-Lii Leu
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan 333, Taiwan; Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Chih-Ching Wu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Department of Otolaryngology - Head & Neck Surgery, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Cheng-Yu Lin
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chang-Yu Pan
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Yung-Fong Tsai
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Department of Anaesthesiology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan 333, Taiwan; Department of Anaesthesiology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; Research Center for Chinese Herbal Medicine, Research Center for Food and Cosmetic Safety, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan; Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City 243, Taiwan.
| |
Collapse
|
16
|
Moldogazieva NT, Lutsenko SV, Terentiev AA. Reactive Oxygen and Nitrogen Species-Induced Protein Modifications: Implication in Carcinogenesis and Anticancer Therapy. Cancer Res 2018; 78:6040-6047. [PMID: 30327380 DOI: 10.1158/0008-5472.can-18-0980] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 07/23/2018] [Accepted: 08/23/2018] [Indexed: 11/16/2022]
Abstract
Cancer is a complex disorder extremely dependent on its microenvironment and highly regulated by multiple intracellular and extracellular stimuli. Studies show that reactive oxygen and nitrogen species (RONS) play key roles in cancer initiation and progression. Accumulation of RONS caused by imbalance between RONS generation and activity of antioxidant system (AOS) has been observed in many cancer types. This leads to alterations in gene expression levels, signal transduction pathways, and protein quality control machinery, that is, processes that regulate cancer cell proliferation, migration, invasion, and apoptosis. This review focuses on the latest advancements evidencing that RONS-induced modifications of key redox-sensitive residues in regulatory proteins, that is, cysteine oxidation/S-sulfenylation/S-glutathionylation/S-nitrosylation and tyrosine nitration, represent important molecular mechanisms underlying carcinogenesis. The oxidative/nitrosative modifications cause alterations in activities of intracellular effectors of MAPK- and PI3K/Akt-mediated signaling pathways, transcription factors (Nrf2, AP-1, NFκB, STAT3, and p53), components of ubiquitin/proteasomal and autophagy/lysosomal protein degradation systems, molecular chaperones, and cytoskeletal proteins. Redox-sensitive proteins, RONS-generating enzymes, and AOS components can serve as targets for relevant anticancer drugs. Chemotherapeutic agents exert their action via RONS generation and induction of cancer cell apoptosis, while drug resistance associates with RONS-induced cancer cell survival; this is exploited in selective anticancer therapy strategies. Cancer Res; 78(21); 6040-7. ©2018 AACR.
Collapse
Affiliation(s)
- Nurbubu T Moldogazieva
- Department of Biotechnology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.
| | - Sergey V Lutsenko
- Department of Biotechnology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Alexander A Terentiev
- Department of Biochemistry and Molecular Biology, N.I. Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
17
|
Saletti R, Reina S, Pittalà MG, Magrì A, Cunsolo V, Foti S, De Pinto V. Post-translational modifications of VDAC1 and VDAC2 cysteines from rat liver mitochondria. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2018; 1859:806-816. [DOI: 10.1016/j.bbabio.2018.06.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 06/04/2018] [Accepted: 06/07/2018] [Indexed: 12/14/2022]
|
18
|
Tan DQ, Suda T. Reactive Oxygen Species and Mitochondrial Homeostasis as Regulators of Stem Cell Fate and Function. Antioxid Redox Signal 2018; 29:149-168. [PMID: 28708000 DOI: 10.1089/ars.2017.7273] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
SIGNIFICANCE The precise role and impact of reactive oxygen species (ROS) in stem cells, which are essential for lifelong tissue homeostasis and regeneration, remain of significant interest to the field. The long-term regenerative potential of a stem cell compartment is determined by the delicate balance between quiescence, self-renewal, and differentiation, all of which can be influenced by ROS levels. Recent Advances: The past decade has seen a growing appreciation for the importance of ROS and redox homeostasis in various stem cell compartments, particularly those of hematopoietic, neural, and muscle tissues. In recent years, the importance of proteostasis and mitochondria in relation to stem cell biology and redox homeostasis has garnered considerable interest. CRITICAL ISSUES Here, we explore the reciprocal relationship between ROS and stem cells, with significant emphasis on mitochondria as a core component of redox homeostasis. We discuss how redox signaling, involving cell-fate determining protein kinases and transcription factors, can control stem cell function and fate. We also address the impact of oxidative stress on stem cells, especially oxidative damage of lipids, proteins, and nucleic acids. We further discuss ROS management in stem cells, and present recent evidence supporting the importance of mitochondrial activity and its modulation (via mitochondrial clearance, biogenesis, dynamics, and distribution [i.e., segregation and transfer]) in stem cell redox homeostasis. FUTURE DIRECTIONS Therefore, elucidating the intricate links between mitochondria, cellular metabolism, and redox homeostasis is envisioned to be critical for our understanding of ROS in stem cell biology and its therapeutic relevance in regenerative medicine. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Darren Q Tan
- Cancer Science Institute of Singapore, National University of Singapore , Singapore, Singapore
| | - Toshio Suda
- Cancer Science Institute of Singapore, National University of Singapore , Singapore, Singapore
| |
Collapse
|
19
|
Abstract
Hydrogen peroxide (H2O2) is produced on stimulation of many cell surface receptors and serves as an intracellular messenger in the regulation of diverse physiological events, mostly by oxidizing cysteine residues of effector proteins. Mammalian cells express multiple H2O2-eliminating enzymes, including catalase, glutathione peroxidase (GPx), and peroxiredoxin (Prx). A conserved cysteine in Prx family members is the site of oxidation by H2O2. Peroxiredoxins possess a high-affinity binding site for H2O2 that is lacking in catalase and GPx and which renders the catalytic cysteine highly susceptible to oxidation, with a rate constant several orders of magnitude greater than that for oxidation of cysteine in most H2O2 effector proteins. Moreover, Prxs are abundant and present in all subcellular compartments. The cysteines of most H2O2 effectors are therefore at a competitive disadvantage for reaction with H2O2. Recent Advances: Here we review intracellular sources of H2O2 as well as H2O2 target proteins classified according to biochemical and cellular function. We then highlight two strategies implemented by cells to overcome the kinetic disadvantage of most target proteins with regard to H2O2-mediated oxidation: transient inactivation of local Prx molecules via phosphorylation, and indirect oxidation of target cysteines via oxidized Prx. Critical Issues and Future Directions: Recent studies suggest that only a small fraction of the total pools of Prxs and H2O2 effector proteins localized in specific subcellular compartments participates in H2O2 signaling. Development of sensitive tools to selectively detect phosphorylated Prxs and oxidized effector proteins is needed to provide further insight into H2O2 signaling. Antioxid. Redox Signal. 28, 537-557.
Collapse
Affiliation(s)
- Sue Goo Rhee
- 1 Yonsei Biomedical Research Institute, Yonsei University College of Medicine , Seoul, Korea
| | - Hyun Ae Woo
- 2 College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University , Seoul, Korea
| | - Dongmin Kang
- 3 Department of Life Science, Ewha Womans University , Seoul, Korea
| |
Collapse
|
20
|
Helfenberger KE, Villalba NM, Buchholz B, Boveris A, Poderoso JJ, Gelpi RJ, Poderoso C. Subcellular distribution of ERK phosphorylation in tyrosine and threonine depends on redox status in murine lung cells. PLoS One 2018; 13:e0193022. [PMID: 29489891 PMCID: PMC5831038 DOI: 10.1371/journal.pone.0193022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 02/02/2018] [Indexed: 11/26/2022] Open
Abstract
Activation of ERK1/2 implies the phosphorylation of tyrosine (pTyr) and threonine (pThr) by MEK1/2; both reactions were thought to be cytoplasmic, promoting ERK to reach the nucleus where it activates several transcription factors. In addition, H2O2 concentrations are known to modulate ERK intracellular translocation, which impacts on cellular proliferation. In this context, the objective of this work was to study the sequence of ERK phosphorylation under two redox conditions and to analyze a putative mitochondrial contribution to this process, in LP07 murine lung cells. A time-course of H2O2 administration was used and ERK phosphorylation was analyzed in cytosol, mitochondria and nuclei. At 1μM H2O2, a proliferative redox stimulus, immunoblot revealed a fast and transient increase in cytosol pTyr and a sustained increase in mitochondrial pTyr content. The detection for pThr/pTyrERK (2pERK) showed in cytosol a marked increase at 5 minutes with a fast dephosphorylation after that time, for both H2O2 concentrations. However, at 50 μM H2O2, an anti-proliferative condition, 2pERK was gradually retained in mitochondria. Interestingly, these results were confirmed by in vivo experiments using mice treated with a highly oxidizing agent [H2O2]. By the use of two ERK2 mutant constructions, where Tyr and Thr were replaced by alanine, we confirmed that 2pERK relied almost completely on pThr183. Confocal microscopy confirmed ERK subcellular distribution dependence on the incidence of cytosolic pTyr and mitochondrial pThr at 1μM H2O2. This work shows for the first time, both in vitro and in vivo, an ERK cycle involving a cross-talk between cytosol and mitochondria phosphorylation events, which may play a significant role in cell cycle progression, proliferation or differentiation under two different redox conditions.
Collapse
Affiliation(s)
- Katia E. Helfenberger
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
| | - Nerina M. Villalba
- Universidad de Buenos Aires, Facultad de Medicina, Hospital de Clínicas “José de San Martín”, Laboratorio del Metabolismo del Oxígeno, Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo (INIGEM), Buenos Aires, Argentina
| | - Bruno Buchholz
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, Argentina
| | - Alberto Boveris
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, Argentina
| | - Juan José Poderoso
- Universidad de Buenos Aires, Facultad de Medicina, Hospital de Clínicas “José de San Martín”, Laboratorio del Metabolismo del Oxígeno, Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo (INIGEM), Buenos Aires, Argentina
| | - Ricardo J. Gelpi
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, Argentina
| | - Cecilia Poderoso
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas (INBIOMED), Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
21
|
Zhang S, Zhang X, Wang K, Xu X, Li M, Zhang J, Zhang Y, Hao J, Sun X, Chen Y, Liu X, Chang Y, Jin R, Wu H, Ge Q. Newly Generated CD4 + T Cells Acquire Metabolic Quiescence after Thymic Egress. THE JOURNAL OF IMMUNOLOGY 2017; 200:1064-1077. [PMID: 29288207 DOI: 10.4049/jimmunol.1700721] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 11/27/2017] [Indexed: 12/19/2022]
Abstract
Mature naive T cells circulate through the secondary lymphoid organs in an actively enforced quiescent state. Impaired cell survival and cell functions could be found when T cells have defects in quiescence. One of the key features of T cell quiescence is low basal metabolic activity. It remains unclear at which developmental stage T cells acquire this metabolic quiescence. We compared mitochondria among CD4 single-positive (SP) T cells in the thymus, CD4+ recent thymic emigrants (RTEs), and mature naive T cells in the periphery. The results demonstrate that RTEs and naive T cells had reduced mitochondrial content and mitochondrial reactive oxygen species when compared with SP thymocytes. This downregulation of mitochondria requires T cell egress from the thymus and occurs early after young T cells enter the circulation. Autophagic clearance of mitochondria, but not mitochondria biogenesis or fission/fusion, contributes to mitochondrial downregulation in RTEs. The enhanced apoptosis signal-regulating kinase 1/MAPKs and reduced mechanistic target of rapamycin activities in RTEs relative to SP thymocytes may be involved in this mitochondrial reduction. These results indicate that the gain of metabolic quiescence is one of the important maturation processes during SP-RTE transition. Together with functional maturation, it promotes the survival and full responsiveness to activating stimuli in young T cells.
Collapse
Affiliation(s)
- Shusong Zhang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing 100191, China
| | - Xinwei Zhang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing 100191, China
| | - Ke Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing 100191, China
| | - Xi Xu
- Center for Molecular Metabolism, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Mingyang Li
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing 100191, China
| | - Jun Zhang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing 100191, China
| | - Yan Zhang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing 100191, China
| | - Jie Hao
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing 100191, China
| | - Xiuyuan Sun
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing 100191, China
| | - Yingyu Chen
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.,Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing 100191, China
| | - Xiaohui Liu
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yingjun Chang
- Peking University Institute of Hematology, People's Hospital, Beijing 100044, China; and
| | - Rong Jin
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; .,Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing 100191, China
| | - Hounan Wu
- Peking University Medical and Health Analytical Center, Peking University Health Science Center, Beijing 100191, China
| | - Qing Ge
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; .,Key Laboratory of Medical Immunology, Ministry of Health, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
22
|
Echeverri-Ruiz N, Haynes T, Landers J, Woods J, Gemma MJ, Hughes M, Del Rio-Tsonis K. A biochemical basis for induction of retina regeneration by antioxidants. Dev Biol 2017; 433:394-403. [PMID: 29291983 PMCID: PMC5753421 DOI: 10.1016/j.ydbio.2017.08.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 08/04/2017] [Accepted: 08/09/2017] [Indexed: 12/19/2022]
Abstract
The use of antioxidants in tissue regeneration has been studied, but their mechanism of action is not well understood. Here, we analyze the role of the antioxidant N-acetylcysteine (NAC) in retina regeneration. Embryonic chicks are able to regenerate their retina after its complete removal from retinal stem/progenitor cells present in the ciliary margin (CM) of the eye only if a source of exogenous factors, such as FGF2, is present. This study shows that NAC modifies the redox status of the CM, initiates self-renewal of the stem/progenitor cells, and induces regeneration in the absence of FGF2. NAC works as an antioxidant by scavenging free radicals either independently or through the synthesis of glutathione (GSH), and/or by reducing oxidized proteins through a thiol disulfide exchange activity. We dissected the mechanism used by NAC to induce regeneration through the use of inhibitors of GSH synthesis and the use of other antioxidants with different biochemical structures and modes of action, and found that NAC induces regeneration through its thiol disulfide exchange activity. Thus, our results provide, for the first time, a biochemical basis for induction of retina regeneration. Furthermore, NAC induction was independent of FGF receptor signaling, but dependent on the MAPK (pErk1/2) pathway.
Collapse
Affiliation(s)
- Nancy Echeverri-Ruiz
- Department of Biology and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA
| | - Tracy Haynes
- Department of Biology and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA
| | - Joseph Landers
- Department of Biology and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA
| | - Justin Woods
- Department of Biology and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA
| | - Michael J Gemma
- Department of Biology and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA
| | - Michael Hughes
- Department of Statistics and Statistical Consulting Center, Miami University, Oxford, OH 45056, USA
| | - Katia Del Rio-Tsonis
- Department of Biology and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA.
| |
Collapse
|
23
|
Keyes JD, Parsonage D, Yammani RD, Rogers LC, Kesty C, Furdui CM, Nelson KJ, Poole LB. Endogenous, regulatory cysteine sulfenylation of ERK kinases in response to proliferative signals. Free Radic Biol Med 2017; 112:534-543. [PMID: 28843779 PMCID: PMC5623068 DOI: 10.1016/j.freeradbiomed.2017.08.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 08/11/2017] [Accepted: 08/22/2017] [Indexed: 01/04/2023]
Abstract
ERK-dependent signaling is key to many pathways through which extracellular signals are transduced into cell-fate decisions. One conundrum is the way in which disparate signals induce specific responses through a common, ERK-dependent kinase cascade. While studies have revealed intricate ways of controlling ERK signaling through spatiotemporal localization and phosphorylation dynamics, additional modes of ERK regulation undoubtedly remain to be discovered. We hypothesized that fine-tuning of ERK signaling could occur by cysteine oxidation. We report that ERK is actively and directly oxidized by signal-generated H2O2 during proliferative signaling, and that ERK oxidation occurs downstream of a variety of receptor classes tested in four cell lines. Furthermore, within the tested cell lines and proliferative signals, we observed that both activation loop-phosphorylated and non-phosphorylated ERK undergo sulfenylation in cells and that dynamics of ERK sulfenylation is dependent on the cell growth conditions prior to stimulation. We also tested the effect of endogenous ERK oxidation on kinase activity and report that phosphotransfer reactions are reversibly inhibited by oxidation by as much as 80-90%, underscoring the importance of considering this additional modification when assessing ERK activation in response to extracellular signals.
Collapse
Affiliation(s)
- Jeremiah D Keyes
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Center for Molecular Signaling, Wake Forest University, USA; Center for Redox Biology and Medicine, Wake Forest School of Medicine, USA
| | - Derek Parsonage
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Center for Redox Biology and Medicine, Wake Forest School of Medicine, USA
| | - Rama D Yammani
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Center for Redox Biology and Medicine, Wake Forest School of Medicine, USA
| | - LeAnn C Rogers
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Center for Molecular Signaling, Wake Forest University, USA
| | - Chelsea Kesty
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Center for Molecular Signaling, Wake Forest University, USA
| | - Cristina M Furdui
- Center for Molecular Signaling, Wake Forest University, USA; Center for Redox Biology and Medicine, Wake Forest School of Medicine, USA; Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Kimberly J Nelson
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Center for Molecular Signaling, Wake Forest University, USA; Center for Redox Biology and Medicine, Wake Forest School of Medicine, USA
| | - Leslie B Poole
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Center for Molecular Signaling, Wake Forest University, USA; Center for Redox Biology and Medicine, Wake Forest School of Medicine, USA.
| |
Collapse
|
24
|
Mistry RK, Brewer AC. Redox regulation of gasotransmission in the vascular system: A focus on angiogenesis. Free Radic Biol Med 2017; 108:500-516. [PMID: 28433660 PMCID: PMC5698259 DOI: 10.1016/j.freeradbiomed.2017.04.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/15/2017] [Accepted: 04/18/2017] [Indexed: 02/06/2023]
Abstract
Reactive oxygen species have emerged as key participants in a broad range of physiological and pathophysiological processes, not least within the vascular system. Diverse cellular functions which have been attributed to some of these pro-oxidants within the vasculature include the regulation of blood pressure, neovascularisation and vascular inflammation. We here highlight the emerging roles of the enzymatically-generated reaction oxygen species, O2- and H2O2, in the regulation of the functions of the gaseous signalling molecules: nitric oxide (NO), carbon monoxide (CO), and hydrogen sulphide (H2S). These gasotransmitters are produced on demand from distinct enzymatic sources and in recent years it has become apparent that they are capable of mediating a number of homeostatic processes within the cardiovascular system including enhanced vasodilation, angiogenesis, wound healing and improved cardiac function following myocardial infarction. In common with O2- and/or H2O2 they signal by altering the functions of target proteins, either by the covalent modification of thiol groups or by direct binding to metal centres within metalloproteins, most notably haem proteins. The regulation of the enzymes which generate NO, CO and H2S have been shown to be influenced at both the transcriptional and post-translational levels by redox-dependent mechanisms, while the activity and bioavailability of the gasotransmitters themselves are also subject to oxidative modification. Within vascular cells, the family of nicotinamide adenine dinucleotide phosphate oxidases (NAPDH oxidases/Noxs) have emerged as functionally significant sources of regulated O2- and H2O2 production and accordingly, direct associations between Nox-generated oxidants and the functions of specific gasotransmitters are beginning to be identified. This review focuses on the current knowledge of the redox-dependent mechanisms which regulate the generation and activity of these gases, with particular reference to their roles in angiogenesis.
Collapse
Affiliation(s)
- Rajesh K Mistry
- Cardiovascular Division, James Black Centre, King's College London BHF Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Alison C Brewer
- Cardiovascular Division, James Black Centre, King's College London BHF Centre of Excellence, 125 Coldharbour Lane, London SE5 9NU, UK.
| |
Collapse
|
25
|
Li R, Zhang LM, Sun WB. RETRACTED: Erythropoietin rescues primary rat cortical neurons from pyroptosis and apoptosis via Erk1/2-Nrf2/Bach1 signal pathway. Brain Res Bull 2017; 130:236-244. [DOI: 10.1016/j.brainresbull.2017.01.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 01/11/2017] [Accepted: 01/24/2017] [Indexed: 11/16/2022]
|
26
|
Zhang DX, Zhang LM, Zhao XC, Sun W. Neuroprotective effects of erythropoietin against sevoflurane-induced neuronal apoptosis in primary rat cortical neurons involving the EPOR-Erk1/2-Nrf2/Bach1 signal pathway. Biomed Pharmacother 2017; 87:332-341. [DOI: 10.1016/j.biopha.2016.12.115] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 12/15/2016] [Accepted: 12/27/2016] [Indexed: 12/26/2022] Open
|
27
|
Sevoflurane pre-conditioning increases phosphorylation of Erk1/2 and HO-1 expression via inhibition of mPTP in primary rat cortical neurons exposed to OGD/R. J Neurol Sci 2017; 372:171-177. [DOI: 10.1016/j.jns.2016.11.055] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 11/03/2016] [Accepted: 11/22/2016] [Indexed: 11/18/2022]
|
28
|
Regulation of mitochondrial functions by protein phosphorylation and dephosphorylation. Cell Biosci 2016; 6:25. [PMID: 27087918 PMCID: PMC4832502 DOI: 10.1186/s13578-016-0089-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 04/01/2016] [Indexed: 12/02/2022] Open
Abstract
The mitochondria are double membrane-bound organelles found in most eukaryotic cells. They generate most of the cell’s energy supply of adenosine triphosphate (ATP). Protein phosphorylation and dephosphorylation are critical mechanisms in the regulation of cell signaling networks and are essential for almost all the cellular functions. For many decades, mitochondria were considered autonomous organelles merely functioning to generate energy for cells to survive and proliferate, and were thought to be independent of the cellular signaling networks. Consequently, phosphorylation and dephosphorylation processes of mitochondrial kinases and phosphatases were largely neglected. However, evidence accumulated in recent years on mitochondria-localized kinases/phosphatases has changed this longstanding view. Mitochondria are increasingly recognized as a hub for cell signaling, and many kinases and phosphatases have been reported to localize in mitochondria and play important functions. However, the strength of the evidence on mitochondrial localization and the activities of the reported kinases and phosphatases vary greatly, and the detailed mechanisms on how these kinases/phosphatases translocate to mitochondria, their subsequent function, and the physiological and pathological implications of their localization are still poorly understood. Here, we provide an updated perspective on the recent advancement in this area, with an emphasis on the implications of mitochondrial kinases/phosphatases in cancer and several other diseases.
Collapse
|
29
|
Panieri E, Santoro MM. ROS signaling and redox biology in endothelial cells. Cell Mol Life Sci 2015; 72:3281-303. [PMID: 25972278 PMCID: PMC11113497 DOI: 10.1007/s00018-015-1928-9] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 04/29/2015] [Accepted: 05/07/2015] [Indexed: 12/14/2022]
Abstract
The purpose of this review is to provide an overview of redox mechanisms, sources and antioxidants that control signaling events in ECs. In particular, we describe which molecules are involved in redox signaling and how they influence the relationship between ECs and other vascular component with regard to angiogenesis. Recent and new tools to investigate physiological ROS signaling will be also discussed. Such findings are providing an overview of the ROS biology relevant for endothelial cells in the context of normal and pathological angiogenic conditions.
Collapse
Affiliation(s)
- Emiliano Panieri
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Massimo M. Santoro
- Laboratory of Endothelial Molecular Biology, Vesalius Research Center, VIB, 3000 Leuven, Belgium
- Laboratory of Endothelial Molecular Biology, Department of Oncology, University of Leuven, 3000 Leuven, Belgium
| |
Collapse
|
30
|
Carbone F, Teixeira PC, Braunersreuther V, Mach F, Vuilleumier N, Montecucco F. Pathophysiology and Treatments of Oxidative Injury in Ischemic Stroke: Focus on the Phagocytic NADPH Oxidase 2. Antioxid Redox Signal 2015; 23:460-489. [PMID: 24635113 PMCID: PMC4545676 DOI: 10.1089/ars.2013.5778] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 03/05/2014] [Accepted: 03/16/2014] [Indexed: 12/23/2022]
Abstract
SIGNIFICANCE Phagocytes play a key role in promoting the oxidative stress after ischemic stroke occurrence. The phagocytic NADPH oxidase (NOX) 2 is a membrane-bound enzyme complex involved in the antimicrobial respiratory burst and free radical production in these cells. RECENT ADVANCES Different oxidants have been shown to induce opposite effects on neuronal homeostasis after a stroke. However, several experimental models support the detrimental effects of NOX activity (especially the phagocytic isoform) on brain recovery after stroke. Therapeutic strategies selectively targeting the neurotoxic ROS and increasing neuroprotective oxidants have recently produced promising results. CRITICAL ISSUES NOX2 might promote carotid plaque rupture and stroke occurrence. In addition, NOX2-derived reactive oxygen species (ROS) released by resident and recruited phagocytes enhance cerebral ischemic injury, activating the inflammatory apoptotic pathways. The aim of this review is to update evidence on phagocyte-related oxidative stress, focusing on the role of NOX2 as a potential therapeutic target to reduce ROS-related cerebral injury after stroke. FUTURE DIRECTIONS Radical scavenger compounds (such as Ebselen and Edaravone) are under clinical investigation as a therapeutic approach against stroke. On the other hand, NOX inhibition might represent a promising strategy to prevent the stroke-related injury. Although selective NOX inhibitors are not yet available, nonselective compounds (such as apocynin and fasudil) provided encouraging results in preclinical studies. Whereas additional studies are needed to better evaluate this therapeutic potential in human beings, the development of specific NOX inhibitors (such as monoclonal antibodies, small-molecule inhibitors, or aptamers) might further improve brain recovery after stroke.
Collapse
Affiliation(s)
- Federico Carbone
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, Geneva, Switzerland
- Department of Internal Medicine, University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino–IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Priscila Camillo Teixeira
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Vincent Braunersreuther
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, Geneva, Switzerland
| | - François Mach
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, Geneva, Switzerland
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Fabrizio Montecucco
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, Geneva, Switzerland
- Department of Internal Medicine, University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino–IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
31
|
New Insight into the Role of Reactive Oxygen Species (ROS) in Cellular Signal-Transduction Processes. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 319:221-54. [PMID: 26404470 DOI: 10.1016/bs.ircmb.2015.07.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Reactive oxygen species (ROS) were once considered to be deleterious agents, contributing to a vast range of pathologies. But, now their protective effects are being appreciated. Both their damaging and beneficial effects are initiated when they target distinct molecules and consequently begin functioning as part of complex signal-transduction pathways. The recognition of ROS as signaling mediators has driven a wealth of research into their roles in both normal and pathophysiological states. The present review assesses the relevant recent literature to outline the current perspectives on redox-signaling mechanisms, physiological implications, and therapeutic strategies. This study highlights that a more fundamental knowledge about many aspects of redox signaling will allow better targeting of ROS, which would in turn improve prophylactic and pharmacotherapy for redox-associated diseases.
Collapse
|
32
|
Liew K, Yong PVC, Navaratnam V, Lim YM, Ho ASH. Differential proteomic analysis on the effects of 2-methoxy-1,4-naphthoquinone towards MDA-MB-231 cell line. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2015; 22:517-527. [PMID: 25981917 DOI: 10.1016/j.phymed.2015.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/31/2015] [Accepted: 03/05/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND We have previously reported the anti-metastatic effects of 2-methoxy-1,4-naphthoquinone (MNQ) against MDA-MB-231 cell line. PURPOSE To investigate the molecular mechanism underlying the anti-metastatic effects of MNQ towards MDA-MB-231 cell line via the comparative proteomic approach. STUDY DESIGN/METHODS Differentially expressed proteins in MNQ-treated MDA-MB-231 cells were identified by using two-dimensional gel electrophoresis coupled with tandem mass spectrometry. Proteins and signalling pathways associated with the identified MNQ-altered proteins were studied by using Western blotting. RESULTS Significant modulation of MDA-MB-231 cell proteome was observed upon treatment with MNQ in which the expressions of 19 proteins were found to be downregulated whereas another eight were upregulated (>1.5 fold, p < 0.05). The altered proteins were mainly related to cytoskeletal functions and regulations, mRNA processing, protein modifications and oxidative stress response. Notably, two of the downregulated proteins, protein S100-A4 (S100A4) and laminin-binding protein (RPSA) are known to play key roles in driving metastasis and were verified using Western blotting. Further investigation using Western blotting also revealed that MNQ decreased the activations of pro-metastatic ERK1/2 and NF-κB signalling pathways. Moreover, MNQ was shown to stimulate the expression of the metastatic suppressor, E-cadherin. CONCLUSION This study reports a proposed mechanism by which MNQ exerts its anti-metastatic effects against MDA-MB-231 cell line. The findings from this study offer new insights on the potential of MNQ to be developed as a novel anti-metastatic agent.
Collapse
Affiliation(s)
- Kitson Liew
- School of Biosciences, Taylor's University, No.1 Jalan Taylor's, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Phelim Voon Chen Yong
- School of Biosciences, Taylor's University, No.1 Jalan Taylor's, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Visweswaran Navaratnam
- School of Biosciences, Taylor's University, No.1 Jalan Taylor's, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Yang Mooi Lim
- Department of Pre-Clinical Sciences, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Lot PT21144, Jalan Sungai Long, Bandar Sungai Long, 43000 Kajang, Selangor Darul Ehsan, Malaysia.
| | - Anthony Siong Hock Ho
- School of Biosciences, Taylor's University, No.1 Jalan Taylor's, 47500 Subang Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
33
|
Bai H, Zhang W, Qin XJ, Zhang T, Wu H, Liu JZ, Hai CX. Hydrogen peroxide modulates the proliferation/quiescence switch in the liver during embryonic development and posthepatectomy regeneration. Antioxid Redox Signal 2015; 22:921-37. [PMID: 25621814 DOI: 10.1089/ars.2014.5960] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
AIMS The liver undergoes marked changes in the rate of proliferation during normal development and regeneration through the coordinated activity of numerous signaling pathways. Little is known, however, about the events that act upstream of these signaling pathways. Here, we explore the modulatory effects of hydrogen peroxide (H2O2) on these pathways in the context of liver development and regeneration. RESULTS We show that H2O2 production during liver development and after partial hepatectomy is tightly regulated in time by specific H2O2-producing and scavenging proteins and dose dependently triggers two distinct pathways. Sustained elevated H2O2 levels are required for the activation of ERK signaling and trigger a shift from quiescence to proliferation. Contrastingly, sustained decreased H2O2 levels are required for the activation of p38 signaling and trigger a shift from proliferation to quiescence. Both events impact the cyclin D and Rb pathways and are involved in liver development and regeneration. Pharmacological lowering of H2O2 levels reduces the extent of fetal hepatocyte proliferation and delays the onset of liver regeneration. Chemical augmentation of H2O2 levels in adult hepatocytes triggers proliferation and delays the termination of liver regeneration. INNOVATION Our results challenge the traditional view of H2O2 as a deleterious stressor in response to liver damage and identify a novel role of endogenous H2O2 in liver development and regeneration. CONCLUSIONS Endogenous H2O2 production is tightly regulated during liver development and regeneration. H2O2 constitutes an important trigger for the proliferation and quiescence transition in hepatocytes via the concentration-dependent activation of the ERK or p38 pathway.
Collapse
Affiliation(s)
- Hua Bai
- Department of Toxicology, Shaanxi Provincial Key Laboratory of Free Radical Biology and Medicine, The Ministry of Education Key Laboratory of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University , Xi'an, China
| | | | | | | | | | | | | |
Collapse
|
34
|
Lampard GR, Wengier DL, Bergmann DC. Manipulation of mitogen-activated protein kinase kinase signaling in the Arabidopsis stomatal lineage reveals motifs that contribute to protein localization and signaling specificity. THE PLANT CELL 2014; 26:3358-71. [PMID: 25172143 PMCID: PMC4371834 DOI: 10.1105/tpc.114.127415] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Revised: 07/15/2014] [Accepted: 08/12/2014] [Indexed: 05/18/2023]
Abstract
When multiple mitogen-activated protein kinase (MAPK) components are recruited recurrently to transduce signals of different origins, and often opposing outcomes, mechanisms to enforce signaling specificity are of utmost importance. These mechanisms are largely uncharacterized in plant MAPK signaling networks. The Arabidopsis thaliana stomatal lineage was previously used to show that when rendered constitutively active, four MAPK kinases (MKKs), MKK4/5/7/9, are capable of perturbing stomatal development and that these kinases comprise two pairs, MKK4/5 and MKK7/9, with both overlapping and divergent functions. We characterized the contributions of specific structural domains of these four "stomatal" MKKs to MAPK signaling output and specificity both in vitro and in vivo within the three discrete cell types of the stomatal lineage. These results verify the influence of functional docking (D) domains of MKKs on MAPK signal output and identify novel regulatory functions for previously uncharacterized structures within the N termini of MKK4/5. Beyond this, we present a novel function of the D-domains of MKK7/9 in regulating the subcellular localization of these kinases. These results provide tools to broadly assess the extent to which these and additional motifs within MKKs function to regulate MAPK signal output throughout the plant.
Collapse
Affiliation(s)
- Gregory R Lampard
- Howard Hughes Medical Institute, Stanford University, Stanford, California 94305-5020
| | - Diego L Wengier
- Howard Hughes Medical Institute, Stanford University, Stanford, California 94305-5020
| | - Dominique C Bergmann
- Howard Hughes Medical Institute, Stanford University, Stanford, California 94305-5020 Department of Biology, Stanford University, Stanford, California 94305-5020
| |
Collapse
|
35
|
Javadov S, Jang S, Agostini B. Crosstalk between mitogen-activated protein kinases and mitochondria in cardiac diseases: therapeutic perspectives. Pharmacol Ther 2014; 144:202-25. [PMID: 24924700 DOI: 10.1016/j.pharmthera.2014.05.013] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 05/30/2014] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases cause more mortality and morbidity worldwide than any other diseases. Although many intracellular signaling pathways influence cardiac physiology and pathology, the mitogen-activated protein kinase (MAPK) family has garnered significant attention because of its vast implications in signaling and crosstalk with other signaling networks. The extensively studied MAPKs ERK1/2, p38, JNK, and ERK5, demonstrate unique intracellular signaling mechanisms, responding to a myriad of mitogens and stressors and influencing the signaling of cardiac development, metabolism, performance, and pathogenesis. Definitive relationships between MAPK signaling and cardiac dysfunction remain elusive, despite 30 years of extensive clinical studies and basic research of various animal/cell models, severities of stress, and types of stimuli. Still, several studies have proven the importance of MAPK crosstalk with mitochondria, powerhouses of the cell that provide over 80% of ATP for normal cardiomyocyte function and play a crucial role in cell death. Although many questions remain unanswered, there exists enough evidence to consider the possibility of targeting MAPK-mitochondria interactions in the prevention and treatment of heart disease. The goal of this review is to integrate previous studies into a discussion of MAPKs and MAPK-mitochondria signaling in cardiac diseases, such as myocardial infarction (ischemia), hypertrophy and heart failure. A comprehensive understanding of relevant molecular mechanisms, as well as challenges for studies in this area, will facilitate the development of new pharmacological agents and genetic manipulations for therapy of cardiovascular diseases.
Collapse
Affiliation(s)
- Sabzali Javadov
- Department of Physiology, School of Medicine, University of Puerto Rico, PR, USA.
| | - Sehwan Jang
- Department of Physiology, School of Medicine, University of Puerto Rico, PR, USA
| | - Bryan Agostini
- Department of Physiology, School of Medicine, University of Puerto Rico, PR, USA
| |
Collapse
|
36
|
Ginsenoside Rd attenuates mitochondrial permeability transition and cytochrome C release in isolated spinal cord mitochondria: involvement of kinase-mediated pathways. Int J Mol Sci 2014; 15:9859-77. [PMID: 24897022 PMCID: PMC4100126 DOI: 10.3390/ijms15069859] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 05/08/2014] [Accepted: 05/21/2014] [Indexed: 12/17/2022] Open
Abstract
Ginsenoside Rd (Rd), one of the main active ingredients in Panax ginseng, has multifunctional activity via different mechanisms and neuroprotective effects that are exerted probably via its antioxidant or free radical scavenger action. However, the effects of Rd on spinal cord mitochondrial dysfunction and underlying mechanisms are still obscure. In this study, we sought to investigate the in vitro effects of Rd on mitochondrial integrity and redox balance in isolated spinal cord mitochondria. We verified that Ca2+ dissipated the membrane potential, provoked mitochondrial swelling and decreased NAD(P)H matrix content, which were all attenuated by Rd pretreatment in a dose-dependent manner. In contrast, Rd was not able to inhibit Ca2+ induced mitochondrial hydrogen peroxide generation. The results of Western blot showed that Rd significantly increased the expression of p-Akt and p-ERK, but had no effects on phosphorylation of PKC and p38. In addition, Rd treatment significantly attenuated Ca2+ induced cytochrome c release, which was partly reversed by antagonists of Akt and ERK, but not p-38 inhibitor. The effects of bisindolylmaleimide, a PKC inhibitor, on Rd-induced inhibition of cytochrome c release seem to be at the level of its own detrimental activity on mitochondrial function. Furthermore, we also found that pretreatment with Rd in vivo (10 and 50 mg/kg) protected spinal cord mitochondria against Ca2+ induced mitochondrial membrane potential dissipation and cytochrome c release. It is concluded that Rd regulate mitochondrial permeability transition pore formation and cytochrome c release through protein kinases dependent mechanism involving activation of intramitochondrial Akt and ERK pathways.
Collapse
|
37
|
Hernández-Reséndiz S, Zazueta C. PHO-ERK1/2 interaction with mitochondria regulates the permeability transition pore in cardioprotective signaling. Life Sci 2014; 108:13-21. [PMID: 24835217 DOI: 10.1016/j.lfs.2014.04.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 04/03/2014] [Accepted: 04/30/2014] [Indexed: 12/24/2022]
Abstract
AIMS The molecular mechanism(s) by which extracellular signal-regulated kinase 1/2 (ERK1/2) and other kinases communicate with downstream targets have not been fully determined. Multiprotein signaling complexes undergoing spatiotemporal redistribution may enhance their interaction with effector proteins promoting cardioprotective response. Particularly, it has been proposed that some active kinases in association with caveolae may converge into mitochondria. Therefore, in this study we investigate if PHO-ERK1/2 interaction with mitochondria may provide a mechanistic link in the regulation of these organelles in cardioprotective signaling. MAIN METHODS Using a model of dilated cardiomyopathy followed by ischemia-reperfusion injury, we determined ERK1/2 signaling at the level of mitochondria and evaluated its effect on the permeability transition pore. KEY FINDINGS The most important finding of the present study is that, under cardioprotective conditions, a subpopulation of activated ERK1/2 was directed to the mitochondrial membranes through vesicular trafficking, concurring with increased phosphorylation of mitochondrial proteins and inhibition of the mitochondrial permeability transition pore opening. In addition, our results suggest that vesicles enriched with caveolin-3 could form structures that may drive ERK1/2, GSK3β and Akt to mitochondria. SIGNIFICANCE Signaling complexes including PHO-ERK, PHO-Akt, PHO-eNOS and caveolin-3 contribute to cardioprotection by directly targeting the mitochondrial proteome and regulating the opening of the permeability transition pore in this model.
Collapse
Affiliation(s)
- Sauri Hernández-Reséndiz
- Department of Cardiovascular Biomedicine, National Institute of Cardiology, I.Ch., Juan Badiano No. 1, Colonia Sección XVI, Mexico 14080, DF, Mexico
| | - Cecilia Zazueta
- Department of Cardiovascular Biomedicine, National Institute of Cardiology, I.Ch., Juan Badiano No. 1, Colonia Sección XVI, Mexico 14080, DF, Mexico.
| |
Collapse
|
38
|
Dingley SD, Polyak E, Ostrovsky J, Srinivasan S, Lee I, Rosenfeld AB, Tsukikawa M, Xiao R, Selak MA, Coon JJ, Hebert AS, Grimsrud PA, Kwon YJ, Pagliarini DJ, Gai X, Schurr TG, Hüttemann M, Nakamaru-Ogiso E, Falk MJ. Mitochondrial DNA variant in COX1 subunit significantly alters energy metabolism of geographically divergent wild isolates in Caenorhabditis elegans. J Mol Biol 2014; 426:2199-216. [PMID: 24534730 DOI: 10.1016/j.jmb.2014.02.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 02/05/2014] [Accepted: 02/06/2014] [Indexed: 12/12/2022]
Abstract
Mitochondrial DNA (mtDNA) sequence variation can influence the penetrance of complex diseases and climatic adaptation. While studies in geographically defined human populations suggest that mtDNA mutations become fixed when they have conferred metabolic capabilities optimally suited for a specific environment, it has been challenging to definitively assign adaptive functions to specific mtDNA sequence variants in mammals. We investigated whether mtDNA genome variation functionally influences Caenorhabditis elegans wild isolates of distinct mtDNA lineages and geographic origins. We found that, relative to N2 (England) wild-type nematodes, CB4856 wild isolates from a warmer native climate (Hawaii) had a unique p.A12S amino acid substitution in the mtDNA-encoded COX1 core catalytic subunit of mitochondrial complex IV (CIV). Relative to N2, CB4856 worms grown at 20°C had significantly increased CIV enzyme activity, mitochondrial matrix oxidant burden, and sensitivity to oxidative stress but had significantly reduced lifespan and mitochondrial membrane potential. Interestingly, mitochondrial membrane potential was significantly increased in CB4856 grown at its native temperature of 25°C. A transmitochondrial cybrid worm strain, chpIR (M, CB4856>N2), was bred as homoplasmic for the CB4856 mtDNA genome in the N2 nuclear background. The cybrid strain also displayed significantly increased CIV activity, demonstrating that this difference results from the mtDNA-encoded p.A12S variant. However, chpIR (M, CB4856>N2) worms had significantly reduced median and maximal lifespan relative to CB4856, which may relate to their nuclear-mtDNA genome mismatch. Overall, these data suggest that C. elegans wild isolates of varying geographic origins may adapt to environmental challenges through mtDNA variation to modulate critical aspects of mitochondrial energy metabolism.
Collapse
Affiliation(s)
- Stephen D Dingley
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Erzsebet Polyak
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Julian Ostrovsky
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Satish Srinivasan
- Department of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Icksoo Lee
- Dankook University College of Medicine, Yongin-si, Gyeonggi-do, South Korea
| | - Amy B Rosenfeld
- Department of Molecular Pharmacology and Therapeutics, Loyola University Health Sciences Division, Maywood, IL, 60153, USA
| | - Mai Tsukikawa
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Rui Xiao
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mary A Selak
- Mitochondria Research Core Facility, The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, 19104, USA
| | - Joshua J Coon
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA; Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA; Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Alexander S Hebert
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA; Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Paul A Grimsrud
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Young Joon Kwon
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| | - David J Pagliarini
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Xiaowu Gai
- Department of Molecular Pharmacology and Therapeutics, Loyola University Health Sciences Division, Maywood, IL, 60153, USA
| | - Theodore G Schurr
- Department of Anthropology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics and Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Eiko Nakamaru-Ogiso
- Department of Biochemistry and Biophysics, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104, USA
| | - Marni J Falk
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
39
|
Specific acetylation of p53 by HDAC inhibition prevents DNA damage-induced apoptosis in neurons. J Neurosci 2013; 33:8621-32. [PMID: 23678107 DOI: 10.1523/jneurosci.5214-12.2013] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Histone deacetylase (HDAC) inhibitors have been used to promote neuronal survival and ameliorate neurological dysfunction in a host of neurodegenerative disease models. The precise molecular mechanisms whereby HDAC inhibitors prevent neuronal death are currently the focus of intensive research. Here we demonstrate that HDAC inhibition prevents DNA damage-induced neurodegeneration by modifying the acetylation pattern of the tumor suppressor p53, which decreases its DNA-binding and transcriptional activation of target genes. Specifically, we identify that acetylation at K382 and K381 prevents p53 from associating with the pro-apoptotic PUMA gene promoter, activating transcription, and inducing apoptosis in mouse primary cortical neurons. Paradoxically, acetylation of p53 at the same lysines in various cancer cell lines leads to the induction of PUMA expression and death. Together, our data provide a molecular understanding of the specific outcomes of HDAC inhibition and suggest that strategies aimed at enhancing p53 acetylation at K381 and K382 might be therapeutically viable for capturing the beneficial effects in the CNS, without compromising tumor suppression.
Collapse
|
40
|
Kathiria AS, Butcher MA, Hansen JM, Theiss AL. Nrf2 is not required for epithelial prohibitin-dependent attenuation of experimental colitis. Am J Physiol Gastrointest Liver Physiol 2013; 304:G885-96. [PMID: 23494124 PMCID: PMC3652068 DOI: 10.1152/ajpgi.00327.2012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Inflammatory bowel disease is associated with increased reactive oxygen species (ROS) and decreased antioxidant response in the intestinal mucosa. Expression of the mitochondrial protein prohibitin (PHB) is also decreased during intestinal inflammation. Our previous study showed that genetic restoration of colonic epithelial PHB expression [villin-PHB transgenic (PHB Tg) mice] attenuated dextran sodium sulfate (DSS)-induced colitis/oxidative stress and sustained expression of colonic nuclear factor erythroid 2-related factor 2 (Nrf2), a cytoprotective transcription factor. This study investigated the role of Nrf2 in mediating PHB-induced protection against colitis and expression of the antioxidant response element (ARE)-regulated antioxidant genes heme oxygenase-1 (HO-1) and NAD(P)H quinone oxidoreductase-1 (NQO-1). PHB-transfected Caco-2-BBE human intestinal epithelial cells maintained increased ARE activation and decreased intracellular ROS levels compared with control vector-transfected cells during Nrf2 knockdown by small interfering RNA. Treatment with the ERK inhibitor PD-98059 decreased PHB-induced ARE activation, suggesting that ERK constitutes a significant portion of PHB-mediated ARE activation in Caco-2-BBE cells. PHB Tg, Nrf2(-/-), and PHB Tg/Nrf2(-/-) mice were treated with DSS or 2,4,6-trinitrobenzene sulfonic acid (TNBS), and inflammation and expression of HO-1 and NQO-1 were assessed. PHB Tg/Nrf2(-/-) mice mimicked PHB Tg mice, with attenuated DSS- or TNBS-induced colitis and induction of colonic HO-1 and NQO-1 expression, despite deletion of Nrf2. PHB Tg/Nrf2(-/-) mice exhibited increased activation of ERK during colitis. Our results suggest that maintaining expression of intestinal epithelial cell PHB, which is decreased during colitis, reduces the severity of inflammation and increases colonic levels of the antioxidants HO-1 and NQO-1 via a mechanism independent of Nrf2.
Collapse
Affiliation(s)
- Arwa S. Kathiria
- 1Division of Gastroenterology, Department of Internal Medicine, Baylor Research Institute, Baylor University Medical Center, Dallas, Texas;
| | - Mackenzie A. Butcher
- 1Division of Gastroenterology, Department of Internal Medicine, Baylor Research Institute, Baylor University Medical Center, Dallas, Texas;
| | - Jason M. Hansen
- 2Division of Pulmonary, Allergy/Immunology, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory School of Medicine, Emory University, Atlanta, Georgia; and ,3Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Arianne L. Theiss
- 1Division of Gastroenterology, Department of Internal Medicine, Baylor Research Institute, Baylor University Medical Center, Dallas, Texas;
| |
Collapse
|
41
|
Fermoselle C, García-Arumí E, Puig-Vilanova E, Andreu AL, Urtreger AJ, de Kier Joffé EDB, Tejedor A, Puente-Maestu L, Barreiro E. Mitochondrial dysfunction and therapeutic approaches in respiratory and limb muscles of cancer cachectic mice. Exp Physiol 2013; 98:1349-65. [PMID: 23625954 DOI: 10.1113/expphysiol.2013.072496] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? We explored whether experimental cancer-induced cachexia may alter mitochondrial respiratory chain (MRC) complexes and oxygen uptake in respiratory and peripheral muscles,and whether signalling pathways, proteasome and oxidative stress influence that process. What is the main finding and what is its importance? In cancer cachectic mice, MRC complexes and oxygen consumption were decreased in the diaphragm and gastrocnemius. Blockade of nuclear factor-κB and mitogen-activated protein kinase actions partly restored the muscle mass and force and corrected the MRC dysfunction,while concomitantly reducing tumour burden. Antioxidants improved mitochondrial oxygen consumption without eliciting effects on the loss of muscle mass and force or the tumour size,whereas bortezomib reduced tumour burden without influencing muscle mass and strength or MRC function. Abnormalities in mitochondrial content, morphology and function have been reported in several muscle-wasting conditions. We specifically explored whether experimental cancer-induced cachexia may alter mitochondrial respiratory chain (MRC) complexes and oxygen uptake in respiratory and peripheral muscles, and whether signalling pathways, proteasomes and oxidative stress may influence that process. We evaluated complex I, II and IV enzyme activities (specific activity assays) and MRC oxygen consumption (polarographic measurements) in diaphragm and gastrocnemius of cachectic mice bearing the LP07 lung tumour, with and without treatment with N-acetylcysteine, bortezomib and nuclear factor-κB (sulfasalazine) and mitogen-activated protein kinases (MAPK, U0126) inhibitors (n = 10 per group for all groups). Whole-body and muscle weights and limb muscle force were also assessed in all rodents at baseline and after 1 month. Compared with control animals, cancer cachectic mice showed a significant reduction in body weight gain, smaller sizes of the diaphragm and gastrocnemius, lower muscle strength, decreased activity of complexes I, II and IV and decreased oxygen consumption in both muscles. Blockade of nuclear factor-κB and MAPK actions restored muscle mass and force and corrected the MRC dysfunction in both muscles, while partly reducing tumour burden. Antioxidants improved mitochondrial oxygen uptake without eliciting significant effects on the loss of muscle mass and force or tumour size, whereas the proteasome inhibitor reduced tumour burden without significantly influencing muscle mass and strength or mitochondrial function. In conclusion, nuclear factor-κB and MAPK signalling pathways modulate muscle mass and performance and MRC function of respiratory and limb muscles in this model of experimental cancer cachexia, thus offering targets for therapeutic intervention.
Collapse
Affiliation(s)
- Clara Fermoselle
- Pulmonology Department, Lung Cancer Group, IMIM-Hospital del Mar, Universitat Pompeu Fabra, Barcelona Biomedical Resarch Park, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Corcoran A, Cotter TG. Redox regulation of protein kinases. FEBS J 2013; 280:1944-65. [PMID: 23461806 DOI: 10.1111/febs.12224] [Citation(s) in RCA: 226] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 02/24/2013] [Accepted: 02/27/2013] [Indexed: 12/30/2022]
Abstract
Reactive oxygen species (ROS) have been long regarded as by-products of a cascade of reactions stemming from cellular oxygen metabolism, which, if they accumulate to toxic levels, can have detrimental effects on cellular biomolecules. However, more recently, the recognition of ROS as mediators of cellular communications has led to their classification as signalling mediators in their own right. The prototypic redox-regulated targets downstream of ROS are the protein tyrosine phosphatases, and the wealth of research that has focused on this area has come to shape our understanding of how redox-signalling contributes to and facilitates protein tyrosine phosphorylation signalling cascades. However, it is becoming increasingly apparent that there is more to this system than simply the negative regulation of protein tyrosine phosphatases. Identification of redox-sensitive kinases such as Src led to the slow emergence of a role for redox regulation of tyrosine kinases. A flow of evidence, which has increased exponentially in recent times as a result of the development of new methods for the detection of oxidative modifications, demonstrates that, by concurrent oxidative activation of tyrosine kinases, ROS fine tune the duration and amplification of the phosphorylation signal. A more thorough understanding of the complex regulatory mechanism of redox-modification will allow targeting of both the production of ROS and their downstream effectors for therapeutic purposes. The present review assesses the most relevant recent literature that demonstrates a role for kinase regulation by oxidation, highlights the most significant findings and proposes future directions for this crucial area of redox biology.
Collapse
Affiliation(s)
- Aoife Corcoran
- Tumour Biology Laboratory, Biochemistry Department, Bioscience Research Institute, University College Cork, Ireland
| | | |
Collapse
|
43
|
Wang YT, Tzeng DW, Wang CY, Hong JY, Yang JL. APE1/Ref-1 prevents oxidative inactivation of ERK for G1-to-S progression following lead acetate exposure. Toxicology 2013; 305:120-9. [PMID: 23370007 DOI: 10.1016/j.tox.2013.01.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 01/15/2013] [Accepted: 01/22/2013] [Indexed: 11/27/2022]
Abstract
Apurinic/apyrimidinic endonuclease 1 (APE1)/redox effector factor-1 is a multifunctional enzyme involved in DNA base excision repair and protein redox regulation. Previously, we have showed that lead acetate (Pb) elicits EGFR activation to initiate the SFK/PKCα/Ras/Raf-1/MKK1/2/ERK signaling cascade functioning against genotoxicity. Here, we explore whether APE1 and reactive oxygen species (ROS) affect ERK signaling and cell cycle progression following Pb exposure. We found that Pb induced APE1 expression and ROS generation in CL3 human lung cancer cells. The Pb-elicited ROS levels and cytotoxicity were further enhanced by introducing small interfering RNA specific for APE1 (siAPE1). E3330, an inhibitor of APE1 redox activity, also augmented the ROS levels and cytotoxicity in Pb-treated cells. Intriguingly, the capability of Pb to activate ERK was abolished under siAPE1 or E3330 co-treatments; conversely, forced expression of APE1 up-regulated the ERK activation by Pb or serum in both Cys65-redox activity dependent and independent manners. Moreover, APE1 formed complex with ERK2, and its redox activity could rescue ERK oxidative inactivation. APE1 redox activity also facilitated the Cyclin D1 expression and G1-to-S progression following Pb exposure. In summary, the results indicate that APE1 is a direct redox regulator of ERK for maintaining the kinase activity to promote cell proliferation.
Collapse
Affiliation(s)
- Yi-Ting Wang
- Molecular Carcinogenesis Laboratory, Institute of Biotechnology & Department of Life Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan
| | | | | | | | | |
Collapse
|
44
|
Mechanistic evaluation of a novel small molecule targeting mitochondria in pancreatic cancer cells. PLoS One 2013; 8:e54346. [PMID: 23349858 PMCID: PMC3549929 DOI: 10.1371/journal.pone.0054346] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 12/12/2012] [Indexed: 12/25/2022] Open
Abstract
Background Pancreatic cancer is one of the deadliest cancers with a 5-year survival rate of 6%. Therapeutic options are very limited and there is an unmet medical need for safe and efficacious treatments. Cancer cell metabolism and mitochondria provide unexplored targets for this disease. We recently identified a novel class of triphenylphosphonium salts, TP compounds, with broad- spectrum anticancer properties. We examined the ability of our prototypical compound TP421– chosen for its fluorescent properties – to inhibit the growth of pancreatic cancer cells and further investigated the molecular mechanisms by which it exerts its anticancer effects. Methodology/Principal Findings TP421 exhibited sub-micromolar IC50 values in all the pancreatic cancer cell lines tested using MTT and colony formation assays. TP421 localized predominantly to mitochondria and induced G0/G1 arrest, ROS accumulation, and activation of several stress-regulated kinases. Caspase and PARP-1 cleavage were observed indicating an apoptotic response while LC3B-II and p62 were accumulated indicating inhibition of autophagy. Furthermore, TP421 induced de-phosphorylation of key signaling molecules involved in FAK mediated adhesion that correlated with inhibition of cell migration. Conclusions/Significance TP421 is a representative compound of a new promising class of mitochondrial-targeted agents useful for pancreatic cancer treatment. Because of their unique mechanism of action and efficacy further development is warranted.
Collapse
|
45
|
Antico Arciuch VG, Elguero ME, Poderoso JJ, Carreras MC. Mitochondrial regulation of cell cycle and proliferation. Antioxid Redox Signal 2012; 16:1150-80. [PMID: 21967640 PMCID: PMC3315176 DOI: 10.1089/ars.2011.4085] [Citation(s) in RCA: 317] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 10/03/2011] [Accepted: 10/03/2011] [Indexed: 01/01/2023]
Abstract
Eukaryotic mitochondria resulted from symbiotic incorporation of α-proteobacteria into ancient archaea species. During evolution, mitochondria lost most of the prokaryotic bacterial genes and only conserved a small fraction including those encoding 13 proteins of the respiratory chain. In this process, many functions were transferred to the host cells, but mitochondria gained a central role in the regulation of cell proliferation and apoptosis, and in the modulation of metabolism; accordingly, defective organelles contribute to cell transformation and cancer, diabetes, and neurodegenerative diseases. Most cell and transcriptional effects of mitochondria depend on the modulation of respiratory rate and on the production of hydrogen peroxide released into the cytosol. The mitochondrial oxidative rate has to remain depressed for cell proliferation; even in the presence of O₂, energy is preferentially obtained from increased glycolysis (Warburg effect). In response to stress signals, traffic of pro- and antiapoptotic mitochondrial proteins in the intermembrane space (B-cell lymphoma-extra large, Bcl-2-associated death promoter, Bcl-2 associated X-protein and cytochrome c) is modulated by the redox condition determined by mitochondrial O₂ utilization and mitochondrial nitric oxide metabolism. In this article, we highlight the traffic of the different canonical signaling pathways to mitochondria and the contributions of organelles to redox regulation of kinases. Finally, we analyze the dynamics of the mitochondrial population in cell cycle and apoptosis.
Collapse
Affiliation(s)
| | - María Eugenia Elguero
- Laboratory of Oxygen Metabolism, University of Buenos Aires, University Hospital, Buenos Aires, Argentina
| | - Juan José Poderoso
- Laboratory of Oxygen Metabolism, University of Buenos Aires, University Hospital, Buenos Aires, Argentina
- Department of Internal Medicine, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
- CONICET, Buenos Aires, Argentina
| | - María Cecilia Carreras
- Laboratory of Oxygen Metabolism, University of Buenos Aires, University Hospital, Buenos Aires, Argentina
- CONICET, Buenos Aires, Argentina
- Department of Clinical Biochemistry, INFIBIOC and School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
46
|
Hristova M, Spiess PC, Kasahara DI, Randall MJ, Deng B, van der Vliet A. The tobacco smoke component, acrolein, suppresses innate macrophage responses by direct alkylation of c-Jun N-terminal kinase. Am J Respir Cell Mol Biol 2012; 46:23-33. [PMID: 21778411 PMCID: PMC3262655 DOI: 10.1165/rcmb.2011-0134oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 07/07/2011] [Indexed: 12/21/2022] Open
Abstract
The respiratory innate immune system is often compromised by tobacco smoke exposure, and previous studies have indicated that acrolein, a reactive electrophile in tobacco smoke, may contribute to the immunosuppressive effects of smoking. Exposure of mice to acrolein at concentrations similar to those in cigarette smoke (5 ppm, 4 h) significantly suppressed alveolar macrophage responses to bacterial LPS, indicated by reduced induction of nitric oxide synthase 2, TNF-α, and IL-12p40. Mechanistic studies with bone marrow-derived macrophages or MH-S macrophages demonstrated that acrolein (1-30 μM) attenuated these LPS-mediated innate responses in association with depletion of cellular glutathione, although glutathione depletion itself was not fully responsible for these immunosuppressive effects. Inhibitory actions of acrolein were most prominent after acute exposure (<2 h), indicating the involvement of direct and reversible interactions of acrolein with critical signaling pathways. Among the key signaling pathways involved in innate macrophage responses, acrolein marginally affected LPS-mediated activation of nuclear factor (NF)-κB, and significantly suppressed phosphorylation of c-Jun N-terminal kinase (JNK) and activation of c-Jun. Using biotin hydrazide labeling, NF-κB RelA and p50, as well as JNK2, a critical mediator of innate macrophage responses, were revealed as direct targets for alkylation by acrolein. Mass spectrometry analysis of acrolein-modified recombinant JNK2 indicated adduction to Cys(41) and Cys(177), putative important sites involved in mitogen-activated protein kinase (MAPK) kinase (MEK) binding and JNK2 phosphorylation. Our findings indicate that direct alkylation of JNK2 by electrophiles, such as acrolein, may be a prominent and hitherto unrecognized mechanism in their immunosuppressive effects, and may be a major factor in smoking-induced effects on the immune system.
Collapse
Affiliation(s)
| | | | | | | | - Bin Deng
- Department of Biology and Proteomics Core Facility, University of Vermont, Burlington, Vermont
| | | |
Collapse
|
47
|
Wortzel I, Seger R. The ERK Cascade: Distinct Functions within Various Subcellular Organelles. Genes Cancer 2011; 2:195-209. [PMID: 21779493 DOI: 10.1177/1947601911407328] [Citation(s) in RCA: 398] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The extracellular signal-regulated kinase 1/2 (ERK1/2) cascade is a central signaling pathway that regulates a wide variety of stimulated cellular processes, including mainly proliferation, differentiation, and survival, but apoptosis and stress response as well. The ability of this linear cascade to induce so many distinct and even opposing effects after various stimulations raises the question as to how the signaling specificity of the cascade is regulated. Over the past years, several specificity-mediating mechanisms have been elucidated, including temporal regulation, scaffolding interactions, crosstalks with other signaling components, substrate competition, and multiple components in each tier of the cascade. In addition, spatial regulation of various components of the cascade is probably one of the main ways by which signals can be directed to some downstream targets and not to others. In this review, we describe first the components of the ERK1/2 cascade and their mode of regulation by kinases, phosphatases, and scaffold proteins. In the second part, we focus on the role of MEK1/2 and ERK1/2 compartmentalization in the nucleus, mitochondria, endosomes, plasma membrane, cytoskeleton, and Golgi apparatus. We explain that this spatial distribution may direct ERK1/2 signals to regulate the organelles' activities. However, it can also direct the activity of the cascade's components to the outer surface of the organelles in order to bring them to close proximity to specific cytoplasmic targets. We conclude that the dynamic localization of the ERK1/2 cascade components is an important regulatory mechanism in determining the signaling specificity of the cascade, and its understanding should shed a new light on the understanding of many stimulus-dependent processes.
Collapse
Affiliation(s)
- Inbal Wortzel
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
48
|
Villalta JI, Galli S, Iacaruso MF, Antico Arciuch VG, Poderoso JJ, Jares-Erijman EA, Pietrasanta LI. New algorithm to determine true colocalization in combination with image restoration and time-lapse confocal microscopy to MAP kinases in mitochondria. PLoS One 2011; 6:e19031. [PMID: 21559502 PMCID: PMC3084741 DOI: 10.1371/journal.pone.0019031] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Accepted: 03/14/2011] [Indexed: 11/19/2022] Open
Abstract
The subcellular localization and physiological functions of biomolecules are closely related and thus it is crucial to precisely determine the distribution of different molecules inside the intracellular structures. This is frequently accomplished by fluorescence microscopy with well-characterized markers and posterior evaluation of the signal colocalization. Rigorous study of colocalization requires statistical analysis of the data, albeit yet no single technique has been established as a standard method. Indeed, the few methods currently available are only accurate in images with particular characteristics. Here, we introduce a new algorithm to automatically obtain the true colocalization between images that is suitable for a wide variety of biological situations. To proceed, the algorithm contemplates the individual contribution of each pixel's fluorescence intensity in a pair of images to the overall Pearsońs correlation and Manders' overlap coefficients. The accuracy and reliability of the algorithm was validated on both simulated and real images that reflected the characteristics of a range of biological samples. We used this algorithm in combination with image restoration by deconvolution and time-lapse confocal microscopy to address the localization of MEK1 in the mitochondria of different cell lines. Appraising the previously described behavior of Akt1 corroborated the reliability of the combined use of these techniques. Together, the present work provides a novel statistical approach to accurately and reliably determine the colocalization in a variety of biological images.
Collapse
Affiliation(s)
- Jorge Ignacio Villalta
- Centro de Microscopías Avanzadas, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Soledad Galli
- Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, CIHIDECAR, CONICET, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - María Florencia Iacaruso
- Centro de Microscopías Avanzadas, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Valeria Gabriela Antico Arciuch
- Laboratory of Oxygen Metabolism, University Hospital “José de San Martín,” Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan José Poderoso
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Laboratory of Oxygen Metabolism, University Hospital “José de San Martín,” Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Elizabeth Andrea Jares-Erijman
- Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, CIHIDECAR, CONICET, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Lía Isabel Pietrasanta
- Centro de Microscopías Avanzadas, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| |
Collapse
|
49
|
Miura T, Tanno M, Sato T. Mitochondrial kinase signalling pathways in myocardial protection from ischaemia/reperfusion-induced necrosis. Cardiovasc Res 2010; 88:7-15. [PMID: 20562423 DOI: 10.1093/cvr/cvq206] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Multiple cardioprotective signal pathways that are activated by ischaemic preconditioning (IPC) and those by IPC mimetics converge on mitochondria. Recent studies have shown that pools of Akt, protein kinase C-ε, extracellular-regulated kinases, glycogen synthase kinase-3beta (GSK-3beta), and hexokinases (HK) I and II, are localized in mitochondria in addition to their pools in the cytosol. Accumulating evidence indicates that such 'mitochondrial protein kinases' receive signals from cytosolic molecules and enhance tolerance of myocytes to injury. Proteomic analyses suggest that these kinases form complexes with each other and with subunit proteins of the mitochondrial permeability transition pore (mPTP). Functional relationships between the protein kinases in mitochondria have not been fully clarified, but GSK-3beta and HKs appear to be at the end of the signal pathways and directly responsible for inhibition of opening of the mPTP and, thus, for myocyte protection from necrosis. In this review, recent findings supporting roles of mitochondrial protein kinases in protection from myocardial necrosis after ischaemia/reperfusion are summarized and discussed.
Collapse
Affiliation(s)
- Tetsuji Miura
- Second Department of Internal Medicine, Sapporo Medical University, School of Medicine, Sapporo, Japan.
| | | | | |
Collapse
|
50
|
Smolková K, Plecitá-Hlavatá L, Bellance N, Benard G, Rossignol R, Ježek P. Waves of gene regulation suppress and then restore oxidative phosphorylation in cancer cells. Int J Biochem Cell Biol 2010; 43:950-68. [PMID: 20460169 DOI: 10.1016/j.biocel.2010.05.003] [Citation(s) in RCA: 171] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Revised: 03/05/2010] [Accepted: 05/04/2010] [Indexed: 12/17/2022]
Abstract
We posit the following hypothesis: Independently of whether malignant tumors are initiated by a fundamental reprogramming of gene expression or seeded by stem cells, "waves" of gene expression that promote metabolic changes occur during carcinogenesis, beginning with oncogene-mediated changes, followed by hypoxia-induced factor (HIF)-mediated gene expression, both resulting in the highly glycolytic "Warburg" phenotype and suppression of mitochondrial biogenesis. Because high proliferation rates in malignancies cause aglycemia and nutrient shortage, the third (second oncogene) "wave" of adaptation stimulates glutaminolysis, which in certain cases partially re-establishes oxidative phosphorylation; this involves the LKB1-AMPK-p53, PI3K-Akt-mTOR axes and MYC dysregulation. Oxidative glutaminolysis serves as an alternative pathway compensating for cellular ATP. Together with anoxic glutaminolysis it provides pyruvate, lactate, and the NADPH pool (alternatively to pentose phosphate pathway). Retrograde signaling from revitalized mitochondria might constitute the fourth "wave" of gene reprogramming. In turn, upon reversal of the two Krebs cycle enzymes, glutaminolysis may partially (transiently) function even during anoxia, thereby further promoting malignancy. The history of the carcinogenic process within each malignant tumor determines the final metabolic phenotype of the selected surviving cells, resulting in distinct cancer bioenergetic phenotypes ranging from the highly glycolytic "classic Warburg" to partial or enhanced oxidative phosphorylation. We discuss the bioenergetically relevant functions of oncogenes, the involvement of mitochondrial biogenesis/degradation in carcinogenesis, the yet unexplained Crabtree effect of instant glucose blockade of respiration, and metabolic signaling stemming from the accumulation of succinate, fumarate, pyruvate, lactate, and oxoglutarate by interfering with prolyl hydroxylase domain enzyme-mediated hydroxylation of HIFα prolines.
Collapse
Affiliation(s)
- Katarína Smolková
- Department of Membrane Transport Biophysics, Institute of Physiology, vvi, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | | | | | | | | |
Collapse
|