1
|
Zhang G, Wei H, Zhao A, Yan X, Zhang X, Gan J, Guo M, Wang J, Zhang F, Jiang Y, Liu X, Yang Z, Jiang X. Mitochondrial DNA leakage: underlying mechanisms and therapeutic implications in neurological disorders. J Neuroinflammation 2025; 22:34. [PMID: 39920753 PMCID: PMC11806845 DOI: 10.1186/s12974-025-03363-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
Mitochondrial dysfunction is a pivotal instigator of neuroinflammation, with mitochondrial DNA (mtDNA) leakage as a critical intermediary. This review delineates the intricate pathways leading to mtDNA release, which include membrane permeabilization, vesicular trafficking, disruption of homeostatic regulation, and abnormalities in mitochondrial dynamics. The escaped mtDNA activates cytosolic DNA sensors, especially cyclic gmp-amp synthase (cGAS) signalling and inflammasome, initiating neuroinflammatory cascades via pathways, exacerbating a spectrum of neurological pathologies. The therapeutic promise of targeting mtDNA leakage is discussed in detail, underscoring the necessity for a multifaceted strategy that encompasses the preservation of mtDNA homeostasis, prevention of membrane leakage, reestablishment of mitochondrial dynamics, and inhibition the activation of cytosolic DNA sensors. Advancing our understanding of the complex interplay between mtDNA leakage and neuroinflammation is imperative for developing precision therapeutic interventions for neurological disorders.
Collapse
Affiliation(s)
- Guangming Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China
| | - Huayuan Wei
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China
| | - Anliu Zhao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China
| | - Xu Yan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China
| | - Xiaolu Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China
| | - Jiali Gan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China
| | - Maojuan Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China
| | - Jie Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China
| | - Fayan Zhang
- Heart Disease Department, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Yifang Jiang
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xinxing Liu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China
| | - Zhen Yang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, China.
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, China.
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, No. 10, Poyang Lake Road, Tuanbo New City West District, Jinghai District, Tianjin, 301617, China.
| |
Collapse
|
2
|
Jemison L, Stahl M, Dash RK, Xie D. VDAC Solvation Free Energy Calculation by a Nonuniform Size Modified Poisson-Boltzmann Ion Channel Model. J Comput Chem 2025; 46:e70003. [PMID: 39722599 DOI: 10.1002/jcc.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/23/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024]
Abstract
Voltage-dependent anion channel (VDAC) is the primary conduit for regulated passage of ions and metabolites into and out of a mitochondrion. Calculating the solvation free energy for VDAC is crucial for understanding its stability, function, and interactions within the cellular environment. In this article, numerical schemes for computing the total solvation free energy for VDAC-comprising electrostatic, ideal gas, and excess free energies plus the nonpolar energy-are developed based on a nonuniform size modified Poisson-Boltzmann ion channel (nuSMPBIC) finite element solver along with tetrahedral meshes for VDAC proteins. The current mesh generation package is also updated to improve mesh quality and accelerate mesh generation. A VDAC Solvation Free Energy Calculation (VSFEC) package is then created by integrating these schemes with the updated mesh package, the nuSMPBIC finite element package, the PDB2PQR package, and the OPM database, as well as one uniform SMPBIC finite element package and one Poisson-Boltzmann ion channel (PBIC) finite element package. With the VSFEC package, many numerical experiments are made using six VDAC proteins, eight ionic solutions containing up to four ionic species, including ATP4- and Ca2+, two reference states, different boundary values, and different permittivity constants. The test results underscore the importance of considering nonuniform ionic size effects to explore the varying patterns of the total solvation free energy, and demonstrate the high performance of the VSFEC package for VDAC solvation free energy calculation.
Collapse
Affiliation(s)
- Liam Jemison
- Department of Mathematical Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, USA
| | - Matthew Stahl
- Department of Mathematical Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, USA
| | - Ranjan K Dash
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Dexuan Xie
- Department of Mathematical Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, USA
| |
Collapse
|
3
|
Arenas Velásquez AM, Patino Linares IA, Gaspers LD, Bartlett PJ, Velasques JM, Netto AVG, Thomas AP, Graminha MAS. The binuclear cyclopalladated complex CP2 is targeting ubiquinol-cytochrome c reductase (complex III) of Leishmania amazonensis. Int J Parasitol Drugs Drug Resist 2024; 27:100574. [PMID: 39746288 PMCID: PMC11748178 DOI: 10.1016/j.ijpddr.2024.100574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/29/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025]
Abstract
Leishmaniasis is a neglected disease that remains with a limited number of drugs available for chemotherapy and has an increased drug resistance that affects treatment outcomes. Metal-based drugs such as cyclopalladated complex [Pd(dmba)(μ-N3)]2 (CP2), a Leishmania topoisomerase IB inhibitor involved in calcium dysregulation and mitochondrial dysfunction of the parasite, had been an alternative to outline the appearance of chemoresistance. To identify new molecular targets and point out possible resistance mechanisms, a CP2-resistant Leishmania amazonensis (LaR) was selected by stepwise exposure to increasing drug pressure until a line capable of growth in 13.3 μM CP2. LaR IC50 value was 52.4 μM (4-fold higher than L. amazonensis-wild type, La). LaR promastigotes were cross-resistant to other DNA topoisomerase I inhibitors (camptothecin) and more susceptible to anti-leishmanial drugs pentamidine and miltefosine. A protective effect on cell viability was observed by pretreating the parasite with Ca2+ channel blockers followed by CP2 in La but not in LaR. Analyses of the cell viability of La and LaR using electron transport chain (ETC) inhibitors demonstrated that La is more sensitive than LaR. The studies of mitochondrial oxygen consumption demonstrated that LaR is less susceptible to complex III (ubiquinol-cytochrome c reductase - CcR) inhibitor, antimycin A (AA). CcR activities of La and LaR were equal for both strains in the absence of CP2 and significantly decreased, 69 % for La and 51 % for LaR, in the presence of CP2. This resistance is attributed to overexpression of CcR, confirmed by the RT-qPCR. CcR inhibition by CP2 leads the parasite to increase the reactive oxygen species (ROS) production, principally in La. Therefore, in this work, we suggested that CcR is the main target of CP2 in the mitochondria, acting to inhibit mitochondria respiratory, and the LaR mutant has increased activity of CcR, which reduces the formation of ROS.
Collapse
Affiliation(s)
- Angela Maria Arenas Velásquez
- São Paulo State University (Unesp), School of Pharmaceutical Sciences, Araraquara, São Paulo, Brazil; Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Irwin Alexander Patino Linares
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Lawrence D Gaspers
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Paula J Bartlett
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| | - Jecika M Velasques
- São Paulo State University (Unesp), Institute of Chemistry, Araraquara, São Paulo, Brazil
| | - Adelino V G Netto
- São Paulo State University (Unesp), Institute of Chemistry, Araraquara, São Paulo, Brazil
| | - Andrew P Thomas
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA.
| | - Marcia A S Graminha
- São Paulo State University (Unesp), School of Pharmaceutical Sciences, Araraquara, São Paulo, Brazil.
| |
Collapse
|
4
|
Xu Y, Tummala SR, Chen X, Vardi N. VDAC in Retinal Health and Disease. Biomolecules 2024; 14:654. [PMID: 38927058 PMCID: PMC11201675 DOI: 10.3390/biom14060654] [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: 04/18/2024] [Revised: 05/22/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
The retina, a tissue of the central nervous system, is vital for vision as its photoreceptors capture light and transform it into electrical signals, which are further processed before they are sent to the brain to be interpreted as images. The retina is unique in that it is continuously exposed to light and has the highest metabolic rate and demand for energy amongst all the tissues in the body. Consequently, the retina is very susceptible to oxidative stress. VDAC, a pore in the outer membrane of mitochondria, shuttles metabolites between mitochondria and the cytosol and normally protects cells from oxidative damage, but when a cell's integrity is greatly compromised it initiates cell death. There are three isoforms of VDAC, and existing evidence indicates that all three are expressed in the retina. However, their precise localization and function in each cell type is unknown. It appears that most retinal cells express substantial amounts of VDAC2 and VDAC3, presumably to protect them from oxidative stress. Photoreceptors express VDAC2, HK2, and PKM2-key proteins in the Warburg pathway that also protect these cells. Consistent with its role in initiating cell death, VDAC is overexpressed in the retinal degenerative diseases retinitis pigmentosa, age related macular degeneration (AMD), and glaucoma. Treatment with antioxidants or inhibiting VDAC oligomerization reduced its expression and improved cell survival. Thus, VDAC may be a promising therapeutic candidate for the treatment of these diseases.
Collapse
Affiliation(s)
- Ying Xu
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (Y.X.); (X.C.)
| | - Shanti R. Tummala
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Xiongmin Chen
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (Y.X.); (X.C.)
| | - Noga Vardi
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
5
|
Xue Q, Ji S, Xu H, Yu S. O-GlcNAcylation: a pro-survival response to acute stress in the cardiovascular and central nervous systems. Eur J Med Res 2024; 29:174. [PMID: 38491477 PMCID: PMC10943874 DOI: 10.1186/s40001-024-01773-z] [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: 08/19/2023] [Accepted: 03/06/2024] [Indexed: 03/18/2024] Open
Abstract
O-GlcNAcylation is a unique monosaccharide modification that is ubiquitously present in numerous nucleoplasmic and mitochondrial proteins. The hexosamine biosynthesis pathway (HBP), which is a key branch of glycolysis, provides the unique sugar donor UDP-GlcNAc for the O-GlcNAc modification. Thus, HBP/O-GlcNAcylation can act as a nutrient sensor to perceive changes in nutrient levels and trigger O-GlcNAc modifications of functional proteins in cellular (patho-)physiology, thereby regulating diverse metabolic processes. An imbalance in O-GlcNAcylation has been shown to be a pathogenic contributor to dysfunction in metabolic diseases, including type 2 diabetes, cancer, and neurodegeneration. However, under acute stress conditions, protein O-GlcNAc modification exhibits rapid and transient upregulation, which is strongly correlated with stress tolerance and cell survival. In this context, we discuss the metabolic, pharmacological and genetic modulation of HBP/O-GlcNAc modification in the biological system, the beneficial role of O-GlcNAcylation in regulating stress tolerance for cardioprotection, and neuroprotection, which is a novel and rapidly growing field. Current evidence suggests that transient activation of the O-GlcNAc modification represents a potent pro-survival signalling pathway and may provide a promising strategy for stress-related disorder therapy.
Collapse
Affiliation(s)
- Qiu Xue
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, 226001, China
- Department of General Surgery, Nantong Tumor Hospital, Nantong Fifth People's Hospital, Affiliated Tumor Hospital of Nantong University, 30 Tongyang North Road, Nantong, 226361, China
| | - Shengtao Ji
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, 226001, China
- Department of Neurology, Affiliated Hospital of Nantong University, Medical School of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Hui Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, 226001, China
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, 399 Century Avenue, Nantong, 226001, China
| | - Shu Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, 226001, China.
| |
Collapse
|
6
|
Rodríguez-Quintero P, Rubio-Osornio M, Uribe E, Moreno W, Marín-Castañeda LA, Morales Z, Portila A, Vázquez D, Rubio C. Exposure to Ozone Downregulates Bcl-2 and Increases Executing Caspases-3 and -8 in the Hippocampus, Frontal Cortex, and Cerebellum of Rats. Cureus 2024; 16:e54546. [PMID: 38516464 PMCID: PMC10956716 DOI: 10.7759/cureus.54546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2024] [Indexed: 03/23/2024] Open
Abstract
Introduction Ozone (O3) is one of the most prevalent atmospheric pollutants, arising from a photochemical reaction between volatile organic compounds (VOC), nitrogen oxides (NOx), and sunlight. O3 triggers oxidative stress, resulting in lipid oxidation, inflammation, alterations in metabolic and cellular signaling, and potentially initiating cell death in vulnerable brain regions. Inflammation and oxidative stress are recognized for their ability to induce cell death, primarily through the apoptosis pathway, involving various proteins that participate in this process via two pathways: intrinsic and extrinsic. Objective This study aims to identify the expression of pro-apoptotic proteins and Bcl-2 in the frontal cortex, cerebellum, and hippocampus of rats exposed to O3 acutely. Methods Two groups of 20 Wistar rodents (250-300 g) were established. The control group (n=10) was exposed to unrestricted polluted air for 12 hours, while the experimental group (n=10) was exposed to 1 ppm of O3. After exposure, the animals were sacrificed for immunofluorescence and Western blot analysis. Using a t-test, the arbitrary units of pro-apoptotic proteins and Bcl-2 were compared between the two groups. Results Significant increases in caspase-8 and caspase-3 activation were found in the O3-exposed group compared to the control group, specifically in the frontal cortex, cerebellum, and hippocampus. Additionally, notable changes in Bcl-2 expression were observed in these brain regions. The TUNEL (terminal deoxynucleotidyl transferase dUTP nick end labeling) assay further indicated significant differences in immunopositivity between the groups in the same areas. However, intrinsic apoptotic proteins such as Bax, VDAC1, and cytochrome-c did not show significant differences between the groups within these structures. Western blot analyses aligned with the immunofluorescence results, showing statistically significant concentrations of caspase-8 in the cerebellum, caspase-3 in the hippocampus, and Bcl-2 in the frontal cortex in the O3 exposed group. Conversely, proteins like Bax, cytochrome-c, and VDAC1 did not exhibit significant differences in all analyzed structures. Conclusions This study demonstrates that acute exposure to 1 ppm of ozone can trigger neuronal apoptosis in the frontal cortex, hippocampus, and cerebellum of rats, primarily through the activation of the extrinsic apoptosis pathway via caspase-8 and caspase-3. Additionally, it causes a reduction in Bcl-2 expression, an essential antiapoptotic protein. Despite not observing the activation of intrinsic pathway proteins like BAX, VDAC, or cytochrome-c, the study suggests that chronic O3 exposure might promote cell death by activating this pathway, requiring further long-term research.
Collapse
Affiliation(s)
- Paola Rodríguez-Quintero
- Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Mexico City, MEX
| | - Moisés Rubio-Osornio
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, Mexico City, MEX
| | - Eric Uribe
- Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Mexico City, MEX
| | - Wilhelm Moreno
- Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Mexico City, MEX
| | - Luis A Marín-Castañeda
- Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Mexico City, MEX
| | - Zayra Morales
- Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Mexico City, MEX
| | - Alonso Portila
- Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Mexico City, MEX
| | - David Vázquez
- Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Mexico City, MEX
| | - Carmen Rubio
- Departamento de Neurofisiología, Instituto Nacional de Neurología y Neurocirugía, Mexico City, MEX
| |
Collapse
|
7
|
Song Y, Leem J, Dhanani M, McKirnan MD, Ichikawa Y, Braza J, Harrington EO, Hammond HK, Roth DM, Patel HH. Impact of blood factors on endothelial cell metabolism and function in two diverse heart failure models. PLoS One 2023; 18:e0281550. [PMID: 36780477 PMCID: PMC9924994 DOI: 10.1371/journal.pone.0281550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 01/25/2023] [Indexed: 02/15/2023] Open
Abstract
Role of blood-based factors in development and progression of heart failure (HF) is poorly characterized. Blood contains factors released during pathophysiological states that may impact cellular function and provide mechanistic insights to HF management. We tested effects of blood from two distinct HF models on cardiac metabolism and identified possible cellular targets of the effects. Blood plasma was obtained from daunorubicin- and myocardial infarction-induced HF rabbits (Dauno-HF and MI-HF) and their controls (Dauno-Control and MI-Control). Effects of plasma on bioenergetics of myocardial tissue from healthy mice and cellular cardiac components were assessed using high-resolution respirometry and Seahorse flux analyzer. Since endothelial cell respiration was profoundly affected by HF plasma, effects of plasma on endothelial cell barrier function and death were further evaluated. Western-blotting and electron microscopy were performed to evaluate mitochondrial proteins and morphology. Brief exposure to HF plasma decreased cardiac tissue respiration. Endothelial cell respiration was most impacted by exposure to HF plasma. Endothelial cell monolayer integrity was decreased by incubation with Dauno-HF plasma. Apoptosis and necrosis were increased in cells incubated with Dauno-HF plasma for 24 h. Down-regulation of voltage-dependent anion-selective channel (VDAC)-1, translocase of outer membrane 20 (Tom20), and mitochondrial fission factor (MFF) in cells exposed to Dauno-HF plasma and mitochondrial signal transducer and activator of transcription 3 (Stat3) and MFF in cells exposed to MI-HF plasma were observed. Mitochondrial structure was disrupted in cells exposed to HF plasma. These findings indicate that endothelial cells and mitochondrial structure and function may be primary target where HF pathology manifests and accelerates. High-throughput blood-based screening of HF may provide innovative ways to advance disease diagnosis and management.
Collapse
Affiliation(s)
- Young Song
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joseph Leem
- Veterans Administration San Diego Healthcare System, San Diego, CA, United States of America
| | - Mehul Dhanani
- Veterans Administration San Diego Healthcare System, San Diego, CA, United States of America
| | - M. Dan McKirnan
- Department of Medicine, UCSD School of Medicine, San Diego, CA, United States of America
| | - Yasuhiro Ichikawa
- Veterans Administration San Diego Healthcare System, San Diego, CA, United States of America
| | - Julie Braza
- Department of Medicine, Brown University and the Vascular Research Laboratory, Providence VA Medical Center, Providence, RI, United States of America
| | - Elizabeth O. Harrington
- Department of Medicine, Brown University and the Vascular Research Laboratory, Providence VA Medical Center, Providence, RI, United States of America
| | - H. Kirk Hammond
- Veterans Administration San Diego Healthcare System, San Diego, CA, United States of America
- Department of Medicine, UCSD School of Medicine, San Diego, CA, United States of America
| | - David M. Roth
- Veterans Administration San Diego Healthcare System, San Diego, CA, United States of America
- Department of Anesthesiology, UCSD School of Medicine, San Diego, CA, United States of America
| | - Hemal H. Patel
- Veterans Administration San Diego Healthcare System, San Diego, CA, United States of America
- Department of Anesthesiology, UCSD School of Medicine, San Diego, CA, United States of America
| |
Collapse
|
8
|
Siddiqui SI, Malik C, Ghosh S. Voltage dependent anion channel and its interaction with N-acetyl-L-Cysteine (NAC) under oxidative stress on planar lipid bilayer. Biochimie 2023; 209:150-160. [PMID: 36780980 DOI: 10.1016/j.biochi.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 12/30/2022] [Accepted: 02/08/2023] [Indexed: 02/13/2023]
Abstract
Mitochondria are the major source of Hydrogen Peroxide (H2O2), a reactive oxygen species, in the cells. The reactive oxygen species generated by the mitochondria oxidize major proteins including Voltage Dependent Anion Channel (VDAC). We were interested to know how the effect of H2O2 is countered by antioxidants present around the mitochondria. N-Acetyl-l-Cysteine (NAC) is a naturally existing antioxidant in the cells. Keeping this in view, the modulatory effect of antioxidant NAC on H2O2 oxidized VDAC has been investigated through in vitro electrophysiological studies. First, the effect of H2O2 and NAC was studied on independently incorporated single-channel VDAC. It was observed that NAC suppresses VDAC conductance with a half-maximal inhibitory concentration (IC50) of ∼1.04 μM. In contrast, H2O2 enhances VDAC conductance. Later, oxidative stress was induced by H2O2 on VDAC increased conductance with half-maximal effective concentration (EC50) of ∼302 nM. An application of 1 μM NAC on H2O2 treated (300 nM) VDAC reversed the effect of oxidation. In the next step, NAC and H2O2 were added in reverse order. When oxidative stress was induced using H2O2, reduction in conductance by NAC was 4.5 ± 0.404 nS. The change in conductance is nearly 6.3%. However, if antioxidant NAC was incubated first followed by H2O2 treatment, the conductance of VDAC was 3.09 ± 0.27 nS. The change in conductance is near 33%. Both H2O2 and NAC also affected various conducting states of VDAC. In-silico studies indicated the binding of NAC at Lysine and Glutamic acid of VDAC. Hence, NAC was found to be effective in protection of VDAC against H2O2-induced oxidative stress due to its strong binding.
Collapse
Affiliation(s)
- Shumaila Iqbal Siddiqui
- Department of Biophysics, Benito Juarez Road, University of Delhi South Campus, New Delhi, 110021, India
| | - Chetan Malik
- Department of Biophysics, Benito Juarez Road, University of Delhi South Campus, New Delhi, 110021, India
| | - Subhendu Ghosh
- Department of Biophysics, Benito Juarez Road, University of Delhi South Campus, New Delhi, 110021, India.
| |
Collapse
|
9
|
Dumbali SP, Wenzel PL. Mitochondrial Permeability Transition in Stem Cells, Development, and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1409:1-22. [PMID: 35739412 DOI: 10.1007/5584_2022_720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The mitochondrial permeability transition (mPT) is a process that permits rapid exchange of small molecules across the inner mitochondrial membrane (IMM) and thus plays a vital role in mitochondrial function and cellular signaling. Formation of the pore that mediates this flux is well-documented in injury and disease but its regulation has also emerged as critical to the fate of stem cells during embryonic development. The precise molecular composition of the mPTP has been enigmatic, with far more genetic studies eliminating molecular candidates than confirming them. Rigorous studies in the recent decade have implicated central involvement of the F1Fo ATP synthase, or complex V of the electron transport chain, and continue to confirm a regulatory role for Cyclophilin D (CypD), encoded by Ppif, in modulating the sensitivity of the pore to opening. A host of endogenous molecules have been shown to trigger flux characteristic of mPT, including positive regulators such as calcium ions, reactive oxygen species, inorganic phosphate, and fatty acids. Conductance of the pore has been described as low or high, and reversibility of pore opening appears to correspond with the relative abundance of negative regulators of mPT such as adenine nucleotides, hydrogen ion, and divalent cations that compete for calcium-binding sites in the mPTP. Current models suggest that distinct pores could be responsible for differing reversibility and conductance depending upon cellular context. Indeed, irreversible propagation of mPT inevitably leads to collapse of transmembrane potential, arrest of ATP synthesis, mitochondrial swelling, and cell death. Future studies should clarify ambiguities in mPTP structure and reveal new roles for mPT in dictating specialized cellular functions beyond cell survival that are tied to mitochondrial fitness including stem cell self-renewal and fate. The focus of this review is to describe contemporary models of the mPTP and highlight how pore activity impacts stem cells and development.
Collapse
Affiliation(s)
- Sandeep P Dumbali
- Department of Integrative Biology & Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Pamela L Wenzel
- Department of Integrative Biology & Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Immunology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
10
|
The Mitochondrial Enzyme 17βHSD10 Modulates Ischemic and Amyloid-β-Induced Stress in Primary Mouse Astrocytes. eNeuro 2022; 9:ENEURO.0040-22.2022. [PMID: 36096650 PMCID: PMC9536859 DOI: 10.1523/eneuro.0040-22.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 08/24/2022] [Accepted: 08/31/2022] [Indexed: 12/15/2022] Open
Abstract
Severe brain metabolic dysfunction and amyloid-β accumulation are key hallmarks of Alzheimer's disease (AD). While astrocytes contribute to both pathologic mechanisms, the role of their mitochondria, which is essential for signaling and maintenance of these processes, has been largely understudied. The current work provides the first direct evidence that the mitochondrial metabolic switch 17β-hydroxysteroid dehydrogenase type 10 (17βHSD10) is expressed and active in murine astrocytes from different brain regions. While it is known that this protein is overexpressed in the brains of AD patients, we found that 17βHSD10 is also upregulated in astrocytes exposed to amyloidogenic and ischemic stress. Importantly, such catalytic overexpression of 17βHSD10 inhibits mitochondrial respiration during increased energy demand. This observation contrasts with what has been found in neuronal and cancer model systems, which suggests astrocyte-specific mechanisms mediated by the protein. Furthermore, the catalytic upregulation of the enzyme exacerbates astrocytic damage, reactive oxygen species (ROS) generation and mitochondrial network alterations during amyloidogenic stress. On the other hand, 17βHSD10 inhibition through AG18051 counters most of these effects. In conclusion, our data represents novel insights into the role of astrocytic mitochondria in metabolic and amyloidogenic stress with implications of 17βHSD10 in multiple neurodegenerative mechanisms.
Collapse
|
11
|
Bryant JD, Lei Y, VanPortfliet JJ, Winters AD, West AP. Assessing Mitochondrial DNA Release into the Cytosol and Subsequent Activation of Innate Immune-related Pathways in Mammalian Cells. Curr Protoc 2022; 2:e372. [PMID: 35175686 PMCID: PMC8986093 DOI: 10.1002/cpz1.372] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Mitochondria have emerged as key drivers of mammalian innate immune responses, functioning as signaling hubs to trigger inflammation and orchestrating metabolic switches required for phagocyte activation. Mitochondria also contain damage-associated molecular patterns (DAMPs), molecules that share similarity with pathogen-associated molecular patterns (PAMPs) and can engage innate immune sensors to drive inflammation. The aberrant release of mitochondrial DAMPs during cellular stress and injury is an increasingly recognized trigger of inflammatory responses in human diseases. Mitochondrial DNA (mtDNA) is a particularly potent DAMP that engages multiple innate immune sensors, although mounting evidence suggests that cytosolic mtDNA is primarily detected via the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway. cGAS and STING are widely expressed in mammalian cells and serve as key regulators of type I interferon and cytokine expression in both infectious and inflammatory diseases. Despite growing roles for the mtDNA-cGAS-STING axis in human disease, assays to quantify mtDNA release into the cytosol and approaches to link mtDNA to cGAS-STING signaling are not standardized, which increases the possibility for experimental artifacts and misinterpretation of data. Here, we present a series of protocols for assaying the release of mtDNA into the cytosol and subsequent activation of innate immune signaling in mammalian cells. We highlight genetic and pharmacological approaches to induce and inhibit mtDNA release from mitochondria. We also describe immunofluorescence microscopy and cellular fractionation assays to visualize morphological changes in mtDNA and quantify mtDNA accumulation in the cytosol. Finally, we include protocols to examine mtDNA-dependent cGAS-STING activation by RT-qPCR and western blotting. These methods can be performed with standard laboratory equipment and are highly adaptable to a wide range of mammalian cell types. They will permit researchers working across the spectrum of biological and biomedical sciences to accurately and reproducibly measure cytosolic mtDNA release and resulting innate immune responses. © 2022 Wiley Periodicals LLC. Basic Protocol 1: siRNA-mediated knockdown of TFAM to induce mtDNA instability, cytosolic release, and activation of the cGAS-STING pathway Alternate Protocol: Pharmacological induction of mtDNA release and cGAS-STING activation using ABT-737 and Q-VD-OPH Basic Protocol 2: Isolation and quantitation of DNA from cytosolic, mitochondrial, and nuclear fractions Basic Protocol 3: Pharmacological inhibition of mtDNA replication and release.
Collapse
Affiliation(s)
- Joshua D. Bryant
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX
- These authors contributed equally
| | - Yuanjiu Lei
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX
- These authors contributed equally
| | - Jordyn J. VanPortfliet
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX
| | - Ashley D. Winters
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX
| | - A. Phillip West
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX
| |
Collapse
|
12
|
Abstract
IκΒα (the protein product of NFKBIA gene) has widely been considered a pro- apoptotic factor due to its ability to inhibit the anti-apoptotic transcription factor NFκB. Our findings indicate that IκΒα also exerts a strong anti-apoptotic activity at the outer mitochondria membrane (OMM). This function we uncovered is distinct from its ability to sequester and inhibit NFκB. IκΒα instead binds to voltage dependent anion channel 1 (VDAC1) and Hexokinase 2 (HK2), stabilizes this complex and prevents mitochondria outer membrane permeabilisation (MOMP) and apoptosis.
Collapse
Affiliation(s)
- Evangelos Pazarentzos
- Helen Diller Family Comprehensive Cancer Center. University of California San Francisco, CA, USA
| |
Collapse
|
13
|
VDAC2 and the BCL-2 family of proteins. Biochem Soc Trans 2021; 49:2787-2795. [PMID: 34913469 PMCID: PMC8786305 DOI: 10.1042/bst20210753] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/21/2021] [Accepted: 11/26/2021] [Indexed: 12/26/2022]
Abstract
The BCL-2 protein family govern whether a cell dies or survives by controlling mitochondrial apoptosis. As dysregulation of mitochondrial apoptosis is a common feature of cancer cells, targeting protein–protein interactions within the BCL-2 protein family is a key strategy to seize control of apoptosis and provide favourable outcomes for cancer patients. Non-BCL-2 family proteins are emerging as novel regulators of apoptosis and are potential drug targets. Voltage dependent anion channel 2 (VDAC2) can regulate apoptosis. However, it is unclear how this occurs at the molecular level, with conflicting evidence in the literature for its role in regulating the BCL-2 effector proteins, BAK and BAX. Notably, VDAC2 is required for efficient BAX-mediated apoptosis, but conversely inhibits BAK-mediated apoptosis. This review focuses on the role of VDAC2 in apoptosis, discussing the current knowledge of the interaction between VDAC2 and BCL-2 family proteins and the recent development of an apoptosis inhibitor that targets the VDAC2–BAK interaction.
Collapse
|
14
|
Suslov AV, Afanasyev MA, Degtyarev PA, Chumachenko PV, Ekta MB, Sukhorukov VN, Khotina VA, Yet SF, Sobenin IA, Postnov AY. Molecular Pathogenesis and the Possible Role of Mitochondrial Heteroplasmy in Thoracic Aortic Aneurysm. Life (Basel) 2021; 11:1395. [PMID: 34947926 PMCID: PMC8709403 DOI: 10.3390/life11121395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/26/2021] [Accepted: 12/07/2021] [Indexed: 12/06/2022] Open
Abstract
Thoracic aortic aneurysm (TAA) is a life-threatening condition associated with high mortality, in which the aortic wall is deformed due to congenital or age-associated pathological changes. The mechanisms of TAA development remain to be studied in detail, and are the subject of active research. In this review, we describe the morphological changes of the aortic wall in TAA. We outline the genetic disorders associated with aortic enlargement and discuss the potential role of mitochondrial pathology, in particular mitochondrial DNA heteroplasmy, in the disease pathogenesis.
Collapse
Affiliation(s)
- A. V. Suslov
- National Medical Research Center of Cardiology, Moscow 121552, Russia; (A.V.S.); (M.A.A.); (P.V.C.); (I.A.S.); (A.Y.P.)
- Department of Human Anatomy, First Moscow State Medical University (Sechenov University), Moscow 119435, Russia;
| | - M. A. Afanasyev
- National Medical Research Center of Cardiology, Moscow 121552, Russia; (A.V.S.); (M.A.A.); (P.V.C.); (I.A.S.); (A.Y.P.)
| | - P. A. Degtyarev
- Department of Human Anatomy, First Moscow State Medical University (Sechenov University), Moscow 119435, Russia;
| | - P. V. Chumachenko
- National Medical Research Center of Cardiology, Moscow 121552, Russia; (A.V.S.); (M.A.A.); (P.V.C.); (I.A.S.); (A.Y.P.)
| | - M. Bagheri Ekta
- Research Institute of Human Morphology, Moscow 117418, Russia; (M.B.E.); (V.A.K.)
| | - V. N. Sukhorukov
- National Medical Research Center of Cardiology, Moscow 121552, Russia; (A.V.S.); (M.A.A.); (P.V.C.); (I.A.S.); (A.Y.P.)
- Research Institute of Human Morphology, Moscow 117418, Russia; (M.B.E.); (V.A.K.)
| | - V. A. Khotina
- Research Institute of Human Morphology, Moscow 117418, Russia; (M.B.E.); (V.A.K.)
- Institute of General Pathology and Pathophysiology, Moscow 125315, Russia
| | - S.-F. Yet
- Institute of Cellular and System Medicine, National Health Research Institutes, 35 Keyan Road, Zhunan Town 35053, Taiwan;
| | - I. A. Sobenin
- National Medical Research Center of Cardiology, Moscow 121552, Russia; (A.V.S.); (M.A.A.); (P.V.C.); (I.A.S.); (A.Y.P.)
| | - A. Yu Postnov
- National Medical Research Center of Cardiology, Moscow 121552, Russia; (A.V.S.); (M.A.A.); (P.V.C.); (I.A.S.); (A.Y.P.)
- Research Institute of Human Morphology, Moscow 117418, Russia; (M.B.E.); (V.A.K.)
| |
Collapse
|
15
|
Wang R, Deng M, Yang C, Yu Q, Zhang L, Zhu Q, Guo X. A Qa-SNARE complex contributes to soybean cyst nematode resistance via regulation of mitochondria-mediated cell death. JOURNAL OF EXPERIMENTAL BOTANY 2021; 72:7145-7162. [PMID: 34165531 DOI: 10.1093/jxb/erab301] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 06/23/2021] [Indexed: 05/27/2023]
Abstract
The resistance to Heterodera glycines 1 (Rhg1) locus is widely used by soybean breeders to reduce yield loss caused by soybean cyst nematode (SCN). α-SNAP (α-soluble NSF attachment protein) within Rhg1 locus contributes to SCN resistance by modulation of cell status at the SCN feeding site; however, the underlying mechanism is largely unclear. Here, we identified an α-SNAP-interacting protein, GmSYP31A, a Qa-SNARE (soluble NSF attachment protein receptor) protein from soybean. Expression of GmSYP31A significantly induced cell death in Nicotiana benthamiana leaves, and co-expression of α-SNAP and GmSYP31A could accelerate cell death. Overexpression of GmSYP31A increased SCN resistance, while silencing or overexpression of a dominant-negative form of GmSYP31A increased SCN sensitivity. GmSYP31A expression also disrupted endoplasmic reticulum-Golgi trafficking, and the exocytosis pathway. Moreover, α-SNAP was also found to interact with GmVDAC1D (voltage-dependent anion channel). The cytotoxicity induced by the expression of GmSYP31A could be relieved either with the addition of an inhibitor of VDAC protein, or by silencing the VDAC gene. Taken together, our data not only demonstrate that α-SNAP works together with GmSYP31A to increase SCN resistance through triggering cell death, but also highlight the unexplored link between the mitochondrial apoptosis pathway and vesicle trafficking.
Collapse
Affiliation(s)
- Rui Wang
- State Key Laboratory of Agricultural Microbiology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Miaomiao Deng
- State Key Laboratory of Agricultural Microbiology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Chao Yang
- State Key Laboratory of Agricultural Microbiology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Qianqian Yu
- State Key Laboratory of Agricultural Microbiology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Lei Zhang
- State Key Laboratory of Agricultural Microbiology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Qun Zhu
- State Key Laboratory of Agricultural Microbiology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Xiaoli Guo
- State Key Laboratory of Agricultural Microbiology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| |
Collapse
|
16
|
Burton NR, Kim P, Backus KM. Photoaffinity labelling strategies for mapping the small molecule-protein interactome. Org Biomol Chem 2021; 19:7792-7809. [PMID: 34549230 PMCID: PMC8489259 DOI: 10.1039/d1ob01353j] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Nearly all FDA approved drugs and bioactive small molecules exert their effects by binding to and modulating proteins. Consequently, understanding how small molecules interact with proteins at an molecular level is a central challenge of modern chemical biology and drug development. Complementary to structure-guided approaches, chemoproteomics has emerged as a method capable of high-throughput identification of proteins covalently bound by small molecules. To profile noncovalent interactions, established chemoproteomic workflows typically incorporate photoreactive moieties into small molecule probes, which enable trapping of small molecule-protein interactions (SMPIs). This strategy, termed photoaffinity labelling (PAL), has been utilized to profile an array of small molecule interactions, including for drugs, lipids, metabolites, and cofactors. Herein we describe the discovery of photocrosslinking chemistries, including a comparison of the strengths and limitations of implementation of each chemotype in chemoproteomic workflows. In addition, we highlight key examples where photoaffinity labelling has enabled target deconvolution and interaction site mapping.
Collapse
Affiliation(s)
- Nikolas R Burton
- Department of Chemistry and Biochemistry, College of Arts and Sciences, UCLA, Los Angeles, CA, 90095, USA.
| | - Phillip Kim
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Keriann M Backus
- Department of Chemistry and Biochemistry, College of Arts and Sciences, UCLA, Los Angeles, CA, 90095, USA.
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| |
Collapse
|
17
|
Loh D, Reiter RJ. Melatonin: Regulation of Biomolecular Condensates in Neurodegenerative Disorders. Antioxidants (Basel) 2021; 10:1483. [PMID: 34573116 PMCID: PMC8465482 DOI: 10.3390/antiox10091483] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022] Open
Abstract
Biomolecular condensates are membraneless organelles (MLOs) that form dynamic, chemically distinct subcellular compartments organizing macromolecules such as proteins, RNA, and DNA in unicellular prokaryotic bacteria and complex eukaryotic cells. Separated from surrounding environments, MLOs in the nucleoplasm, cytoplasm, and mitochondria assemble by liquid-liquid phase separation (LLPS) into transient, non-static, liquid-like droplets that regulate essential molecular functions. LLPS is primarily controlled by post-translational modifications (PTMs) that fine-tune the balance between attractive and repulsive charge states and/or binding motifs of proteins. Aberrant phase separation due to dysregulated membrane lipid rafts and/or PTMs, as well as the absence of adequate hydrotropic small molecules such as ATP, or the presence of specific RNA proteins can cause pathological protein aggregation in neurodegenerative disorders. Melatonin may exert a dominant influence over phase separation in biomolecular condensates by optimizing membrane and MLO interdependent reactions through stabilizing lipid raft domains, reducing line tension, and maintaining negative membrane curvature and fluidity. As a potent antioxidant, melatonin protects cardiolipin and other membrane lipids from peroxidation cascades, supporting protein trafficking, signaling, ion channel activities, and ATPase functionality during condensate coacervation or dissolution. Melatonin may even control condensate LLPS through PTM and balance mRNA- and RNA-binding protein composition by regulating N6-methyladenosine (m6A) modifications. There is currently a lack of pharmaceuticals targeting neurodegenerative disorders via the regulation of phase separation. The potential of melatonin in the modulation of biomolecular condensate in the attenuation of aberrant condensate aggregation in neurodegenerative disorders is discussed in this review.
Collapse
Affiliation(s)
- Doris Loh
- Independent Researcher, Marble Falls, TX 78654, USA
| | - Russel J. Reiter
- Department of Cellular and Structural Biology, UT Health Science Center, San Antonio, TX 78229, USA
| |
Collapse
|
18
|
Shahsavani N, Alizadeh A, Kataria H, Karimi-Abdolrezaee S. Availability of neuregulin-1beta1 protects neurons in spinal cord injury and against glutamate toxicity through caspase dependent and independent mechanisms. Exp Neurol 2021; 345:113817. [PMID: 34314724 DOI: 10.1016/j.expneurol.2021.113817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 07/06/2021] [Accepted: 07/20/2021] [Indexed: 12/27/2022]
Abstract
Spinal cord injury (SCI) causes sensorimotor and autonomic impairment that partly reflects extensive, permanent loss of neurons at the epicenter and penumbra of the injury. Strategies aimed at enhancing neuronal protection are critical to attenuate neurodegeneration and improve neurological recovery after SCI. In rat SCI, we previously uncovered that the tissue levels of neuregulin-1beta 1 (Nrg-1β1) are acutely and persistently downregulated in the injured spinal cord. Nrg-1β1 is well-known for its critical roles in the development, maintenance and physiology of neurons and glia in the developing and adult spinal cord. However, despite this pivotal role, Nrg-1β1 specific effects and mechanisms of action on neuronal injury remain largely unknown in SCI. In the present study, using a clinically-relevant model of compressive/contusive SCI in rats and an in vitro model of glutamate toxicity in primary neurons, we demonstrate Nrg-1β1 provides early neuroprotection through attenuation of reactive oxygen species, lipid peroxidation, necrosis and apoptosis in acute and subacute stages of SCI. Mechanistically, availability of Nrg-1β1 following glutamate challenge protects neurons from caspase-dependent and independent cell death that is mediated by modulation of mitochondria associated apoptotic cascades and MAP kinase and AKT signaling pathways. Altogether, our work provides novel insights into the role and mechanisms of Nrg-1β1 in neuronal injury after SCI and introduces its potential as a new neuroprotective target for this debilitating neurological condition.
Collapse
Affiliation(s)
- Narjes Shahsavani
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Arsalan Alizadeh
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hardeep Kataria
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
19
|
Kim KW, Kim SW, Lim S, Yoo KJ, Hwang KC, Lee S. Neutralization of hexokinase 2-targeting miRNA attenuates the oxidative stress-induced cardiomyocyte apoptosis. Clin Hemorheol Microcirc 2021; 78:57-68. [PMID: 33523042 DOI: 10.3233/ch-200924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hexokinase 2 (HK2) is a metabolic sensor that couples glycolysis and oxidative phosphorylation of mitochondria by binding to the outer mitochondrial membrane (OMM), and it also has been implicated in induction of apoptotic process by regulating the integrity of OMM. When HK2 detaches from the mitochondria, it triggers permeability increase of the OMM and subsequently facilitates the cytosolic release of cytochrome c, a major apoptosis-inducing factor. According to previous studies, a harsh microenvironment created by ischemic heart disease such as low tissue oxygen and nutrients, and increased reactive oxygen species (ROS) can cause cardiomyocyte apoptosis. Under these conditions, the expression of HK2 in heart significantly decrease and such down-regulation of HK2 was correlated to the increased apoptosis of cardiomyocytes. Therefore, prevention of HK2 down-regulation may salvage cardiomyocytes from apoptosis. MicroRNAs are short, non-coding RNAs that either inhibit transcription of target mRNAs or degrade the targeted mRNAs via complementary binding to the 3'UTR (untranslated region) of the targeted mRNAs. Since miRNAs are known to be involved in virtually every biological processes, it is reasonable to assume that the expression of HK2 is also regulated by miRNAs. Currently, to my best knowledge, there is no previous study examined the miRNA-mediated regulation of HK2 in cardiomyocytes. Thus, in the present study, miRNA-mediated modulation of HK2 during ROS (H2O2)-induced cardiomyocyte apoptosis was investigated. First, the expression of HK2 in cardiomyocytes exposed to H2O2 was evaluated. H2O2 (500 μM) induced cardiomyocyte apoptosis and it also decreased the mitochondrial expression of HK2. Based on miRNA-target prediction databases and empirical data, miR-181a was identified as a HK2-targeting miRNA. To further examine the effect of negative regulation of the selected HK2-targeting miRNA on cardiomyocyte apoptosis, anti-miR-181a, which neutralizes endogenous miR-181a, was utilized. Delivery of anti-miR-181a significantly abrogated the H2O2-induced suppression of HK2 expression and subsequent disruption of mitochondrial membrane potential, improving the survival of cardiomyocytes exposed to H2O2. These findings suggest that miR-181a-mediated down-regulation of HK2 contributes to the apoptosis of cardiomyocytes exposed to ROS. Neutralizing miR-181a can be a viable and effective means to prevent cardiomyocyte from apoptosis in ischemic heart disease.
Collapse
Affiliation(s)
- Kwan Wook Kim
- Department of Medicine, The Graduate School, Yonsei University, Seoul, South Korea.,Department of Thoracic and Cardiovascular Surgery, CHA Bundang Medical Center, CHA University, Pangyo, South Korea
| | - Sang Woo Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, South Korea
| | - Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, South Korea
| | - Kyung-Jong Yoo
- Division of Cardiovascular Surgery, Department of Thoracic and Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, South Korea
| | - Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, South Korea
| |
Collapse
|
20
|
VDAC1 as a target in cisplatin anti-tumor activity through promoting mitochondria fusion. Biochem Biophys Res Commun 2021; 560:52-58. [PMID: 33971568 DOI: 10.1016/j.bbrc.2021.04.104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 04/25/2021] [Indexed: 11/24/2022]
Abstract
Cisplatin is one of the most effective anti-cancer drugs, but its efficacy is limited by the development of resistance. Previous studies have shown that mitochondria play critical roles in cisplatin cytotoxicity, however, the exact mechanism of mitochondria involved in cisplatin sensitivity has not been clarified. In this study, cisplatin triggered mitochondrial oxidative stress and the decrease of mitochondria membrane potential in human cervical cancer cells. Then we screened a series of mitochondrial relevant inhibitors, including mitochondrial mPTP inhibitors DIDS and CsA, and mitochondrial respiratory complex inhibitors Rot and TTFA. Among these, only DIDS, as the inhibitor of mitochondrial outer membrane protein VDAC1, showed strong antagonism against cisplatin toxicity. DIDS mitigated cisplatin-induced MFN1-dependent mitochondrial fusion, mitochondrial dysfunction and oxidative damage. These findings demonstrated that VDAC1 may serve as a potential therapeutic target in the increase sensitivity of cisplatin, which provides an attractive pharmacological therapy to improve the effectiveness of chemotherapy.
Collapse
|
21
|
Al-Salam S, Kandhan K, Sudhadevi M. Down regulation of lactate dehydrogenase initiates apoptosis in HeLa and MCF-7 cancer cells through increased voltage-dependent anion channel protein and inhibition of BCL2. Oncotarget 2021; 12:923-935. [PMID: 33953846 PMCID: PMC8092341 DOI: 10.18632/oncotarget.27950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 03/25/2021] [Indexed: 11/25/2022] Open
Abstract
Malignant cells commonly use aerobic glycolysis for ATP production; this is known as the Warburg effect, where pyruvate is converted to lactate, by enzyme lactate dehydrogenase A (LDH-A). In this study, we have investigated the effect of inhibition of LDH-A on cells viability and identifying the mechanism of cell death in HeLa and MCF-7 cancer cells. Human cervical cancer HeLa cell line and breast cancer MCF-7 cell line were used to investigate the effect of inhibition of LDH-A by sodium oxamate on cell survival and proliferation using western blot, spectrophotometry, and immunofluorescent study. There was significant reduction in LDH-A (P < 0.001) and cell viability (P < 0.001) in a dose-dependent mode in both HeLa and MCF-7 SO-treated cancer cells. The voltage-dependent anion channel (VDAC) protein was significantly increased (P < 0.001) in association with decreased LDH-A. The proapoptotic proteins; cytochrome C (P < 0.001), BAX (P < 0.001), cleaved caspase-3 (P < 0.001), cleaved caspase-8 (P < 0.001), and cleaved caspase-9 (P < 0.001) were significantly increased in association with decreased LDH-A. While, the anti-apoptotic protein Bcl2 was significantly decreased (P < 0.001) in association with decreased LDH-A. We conclude that Inhibition of LDH-A can decrease cells viability through activation of intrinsic apoptotic pathway via increased VDAC protein and inhibition of Bcl2 as well as activation of the extrinsic apoptotic pathway through activation of caspase-8.
Collapse
Affiliation(s)
- Suhail Al-Salam
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, AlAin, UAE
| | - Karthishwaran Kandhan
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, AlAin, UAE
| | - Manjusha Sudhadevi
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, AlAin, UAE
| |
Collapse
|
22
|
Abstract
The evolution of the eukaryotic cell from the primal endosymbiotic event involved a complex series of adaptations driven primarily by energy optimization. Transfer of genes from endosymbiont to host and concomitant expansion (by infolding) of the endosymbiont's chemiosmotic membrane greatly increased output of adenosine triphosphate (ATP) and placed selective pressure on the membrane at the host-endosymbiont interface to sustain the energy advantage. It is hypothesized that critical functions at this interface (metabolite exchange, polypeptide import, barrier integrity to proteins and DNA) were managed by a precursor β-barrel protein ("pβB") from which the voltage-dependent anion-selective channel (VDAC) descended. VDAC's role as hub for disparate and increasingly complex processes suggests an adaptability that likely springs from a feature inherited from pβB, retained because of important advantages conferred. It is proposed that this property is the remarkable structural flexibility evidenced in VDAC's gating mechanism, a possible origin of which is discussed.
Collapse
Affiliation(s)
- Carmen A Mannella
- Department of Physiology, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 20201, USA
| |
Collapse
|
23
|
Yang M, Sun J, Stowe DF, Tajkhorshid E, Kwok WM, Camara AKS. Knockout of VDAC1 in H9c2 Cells Promotes Oxidative Stress-Induced Cell Apoptosis through Decreased Mitochondrial Hexokinase II Binding and Enhanced Glycolytic Stress. Cell Physiol Biochem 2021; 54:853-874. [PMID: 32901466 PMCID: PMC7898235 DOI: 10.33594/000000274] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2020] [Indexed: 12/24/2022] Open
Abstract
Background/Aims: The role of VDAC1, the most abundant mitochondrial outer membrane protein, in cell death depends on cell types and stimuli. Both silencing and upregulation of VDAC1 in various type of cancer cell lines can stimulate apoptosis. In contrast, in mouse embryonic stem (MES) cells and mouse embryonic fibroblasts (MEFs), the roles of VDAC1 knockout (VDAC1−/−) in apoptotic cell death are contradictory. The contribution and underlying mechanism of VDAC1−/− in oxidative stress-induced cell death in cardiac cells has not been established. We hypothesized that VDAC1 is an essential regulator of oxidative stress-induced cell death in H9c2 cells. Methods: We knocked out VDAC1 in this rat cardiomyoblast cell line with CRISPR-Cas9 genome editing technique to produce VDAC1−/− H9c2 cells, and determined if VDAC1 is critical in promoting cell death via oxidative stress induced by tert-butylhydroper-oxide (tBHP), an organic peroxide, or rotenone (ROT), an inhibitor of mitochondrial complex I by measuring cell viability with MTT assay, cell death with TUNEL stain and LDH release. The mitochondrial and glycolytic stress were examined by measuring O2 consumption rate (OCR) and extracellular acidification rate (ECAR) with a Seahorse XFp analyzer. Results: We found that under control conditions, VDAC1−/− did not affect H9c2 cell proliferation or mitochondrial respiration. However, compared to the wildtype (WT) cells, exposure to either tBHP or ROT enhanced the production of ROS, ECAR, and the proton (H+) production rate (PPR) from glycolysis, as well as promoted apoptotic cell death in VDAC1−/− H9c2 cells. VDAC1−/− H9c2 cells also exhibited markedly reduced mitochondria-bound hexokinase II (HKII) and Bax. Restoration of VDAC1 in VDAC1−/− H9c2 cells reinstated mitochondria-bound HKII and concomitantly decreased tBHP and ROT-induced ROS production and cell death. Interestingly, mitochondrial respiration remained the same after tBHP treatment in VDAC1−/− and WT H9c2 cells. Conclusion: Our results suggest that VDAC1−/− in H9c2 cells enhances oxidative stress-mediated cell apoptosis that is directly linked to the reduction of mitochondria-bound HKII and concomitantly associated with enhanced ROS production, ECAR, and PPR.
Collapse
Affiliation(s)
- Meiying Yang
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jie Sun
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Institute of Clinical Medicine Research, Department of Gastroenterology and Hepatology, Suzhou Hospital affiliated with Nanjing Medical University, Suzhou, Jiangsu, China
| | - David F Stowe
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Research Service, Zablocki VA Medical Center, Milwaukee, WI, USA.,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Emad Tajkhorshid
- NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana Champaign, Urbana, IL, USA.,Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois at Urbana Champaign, Urbana, IL, USA
| | - Wai-Meng Kwok
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA.,Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Amadou K S Camara
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA, .,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA.,Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
24
|
Duan C, Evison A, Taylor L, Onur S, Morten K, Townley H. The common diabetes drug metformin can diminish the action of citral against Rhabdomyosarcoma cells in vitro. Phytother Res 2020; 35:1378-1388. [PMID: 33280183 DOI: 10.1002/ptr.6898] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/17/2020] [Accepted: 09/20/2020] [Indexed: 01/12/2023]
Abstract
Rhabdomyosarcoma (RMS) is a rare type of soft tissue sarcoma most commonly found in pediatric patients. Despite progress, new and improved drug regimens are needed to increase survival rates. Citral, a natural product plant oil can induce cell death in cancer cells. Another compound, metformin, isolated originally from French lilac and used by diabetics, has been shown to reduce the incidence of cancer in these patients. Application of citral to RMS cells showed increase in cell death, and RD and RH30 cells showed half maximal inhibitory concentration (IC50 ) values as low as 36.28 μM and 62.37 μM, respectively. It was also shown that the citral initiated cell apoptosis through an increase in reactive oxygen species (ROS) and free calcium. In comparison, metformin only showed moderate cell death in RMS cell lines at a very high concentration (1,000 μM). Combinatorial experiments, however, indicated that citral and metformin worked antagonistically when used together. In particular, the ability of metformin to quench the ROS induced by citral could lead to the suppression of activity. These results clearly indicate that while clinical use of citral is a promising anti-tumor therapy, caution should be exercised in patients using metformin for diabetes.
Collapse
Affiliation(s)
- Chengchen Duan
- Nuffield Department of Women's and Reproductive Health, Oxford University, John Radcliffe Hospital, Oxford, UK
| | - Anna Evison
- Nuffield Department of Women's and Reproductive Health, Oxford University, John Radcliffe Hospital, Oxford, UK
| | - Lucy Taylor
- Nuffield Department of Women's and Reproductive Health, Oxford University, John Radcliffe Hospital, Oxford, UK
| | - Simone Onur
- Department of Molecular Prevention, Kiel University, Kiel, Germany
| | - Karl Morten
- Nuffield Department of Women's and Reproductive Health, Oxford University, John Radcliffe Hospital, Oxford, UK
| | - Helen Townley
- Nuffield Department of Women's and Reproductive Health, Oxford University, John Radcliffe Hospital, Oxford, UK.,Department of Engineering Science, Oxford University, Oxford, UK
| |
Collapse
|
25
|
Zhang L, Townsend DM, Morris M, Maldonado EN, Jiang YL, Broome AM, Bethard JR, Ball LE, Tew KD. Voltage-Dependent Anion Channels Influence Cytotoxicity of ME-344, a Therapeutic Isoflavone. J Pharmacol Exp Ther 2020; 374:308-318. [PMID: 32546528 PMCID: PMC7372917 DOI: 10.1124/jpet.120.000009] [Citation(s) in RCA: 3] [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: 03/17/2020] [Accepted: 05/19/2020] [Indexed: 01/27/2023] Open
Abstract
ME-344 is a second-generation cytotoxic isoflavone with anticancer activity promulgated through interference with mitochondrial functions. Using a click chemistry version of the drug together with affinity-enriched mass spectrometry, voltage-dependent anion channels (VDACs) 1 and 2 were identified as drug targets. To determine the importance of VDAC1 or 2 to cytotoxicity, we used lung cancer cells that were either sensitive (H460) or intrinsically resistant (H596) to the drug. In H460 cells, depletion of VDAC1 and VDAC2 by small interfering RNA impacted ME-344 effects by diminishing generation of reactive oxygen species (ROS), preventing mitochondrial membrane potential dissipation, and moderating ME-344-induced cytotoxicity and mitochondrial-mediated apoptosis. Mechanistically, VDAC1 and VDAC2 knockdown prevented ME-344-induced apoptosis by inhibiting Bax mitochondrial translocation and cytochrome c release as well as apoptosis in these H460 cells. We conclude that VDAC1 and 2, as mediators of the response to oxidative stress, have roles in modulating ROS generation, Bax translocation, and cytochrome c release during mitochondrial-mediated apoptosis caused by ME-344. SIGNIFICANCE STATEMENT: Dissecting preclinical drug mechanisms are of significance in development of a drug toward eventual Food and Drug Administration approval.
Collapse
Affiliation(s)
- Leilei Zhang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics (L.Z., M.M., E.N.M., Y.-L.J., A.-M.B., J.R.B., L.E.B., K.D.T.) and Department of Pharmaceutical and Biomedical Sciences (D.M.T.), Medical University of South Carolina, Charleston, South Carolina
| | - Danyelle M Townsend
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics (L.Z., M.M., E.N.M., Y.-L.J., A.-M.B., J.R.B., L.E.B., K.D.T.) and Department of Pharmaceutical and Biomedical Sciences (D.M.T.), Medical University of South Carolina, Charleston, South Carolina
| | - Morgan Morris
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics (L.Z., M.M., E.N.M., Y.-L.J., A.-M.B., J.R.B., L.E.B., K.D.T.) and Department of Pharmaceutical and Biomedical Sciences (D.M.T.), Medical University of South Carolina, Charleston, South Carolina
| | - Eduardo N Maldonado
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics (L.Z., M.M., E.N.M., Y.-L.J., A.-M.B., J.R.B., L.E.B., K.D.T.) and Department of Pharmaceutical and Biomedical Sciences (D.M.T.), Medical University of South Carolina, Charleston, South Carolina
| | - Yu-Lin Jiang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics (L.Z., M.M., E.N.M., Y.-L.J., A.-M.B., J.R.B., L.E.B., K.D.T.) and Department of Pharmaceutical and Biomedical Sciences (D.M.T.), Medical University of South Carolina, Charleston, South Carolina
| | - Ann-Marie Broome
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics (L.Z., M.M., E.N.M., Y.-L.J., A.-M.B., J.R.B., L.E.B., K.D.T.) and Department of Pharmaceutical and Biomedical Sciences (D.M.T.), Medical University of South Carolina, Charleston, South Carolina
| | - Jennifer R Bethard
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics (L.Z., M.M., E.N.M., Y.-L.J., A.-M.B., J.R.B., L.E.B., K.D.T.) and Department of Pharmaceutical and Biomedical Sciences (D.M.T.), Medical University of South Carolina, Charleston, South Carolina
| | - Lauren E Ball
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics (L.Z., M.M., E.N.M., Y.-L.J., A.-M.B., J.R.B., L.E.B., K.D.T.) and Department of Pharmaceutical and Biomedical Sciences (D.M.T.), Medical University of South Carolina, Charleston, South Carolina
| | - Kenneth D Tew
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics (L.Z., M.M., E.N.M., Y.-L.J., A.-M.B., J.R.B., L.E.B., K.D.T.) and Department of Pharmaceutical and Biomedical Sciences (D.M.T.), Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
26
|
Jeremiasse B, Matta C, Fellows CR, Boocock DJ, Smith JR, Liddell S, Lafeber F, van Spil WE, Mobasheri A. Alterations in the chondrocyte surfaceome in response to pro-inflammatory cytokines. BMC Mol Cell Biol 2020; 21:47. [PMID: 32586320 PMCID: PMC7318434 DOI: 10.1186/s12860-020-00288-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 06/03/2020] [Indexed: 02/07/2023] Open
Abstract
Background Chondrocytes are exposed to an inflammatory micro-environment in the extracellular matrix (ECM) of articular cartilage in joint diseases such as osteoarthritis (OA) and rheumatoid arthritis (RA). In OA, degenerative changes and low-grade inflammation within the joint transform the behaviour and metabolism of chondrocytes, disturb the balance between ECM synthesis and degradation, and alter the osmolality and ionic composition of the micro-environment. We hypothesize that chondrocytes adjust their physiology to the inflammatory microenvironment by modulating the expression of cell surface proteins, collectively referred to as the ‘surfaceome’. Therefore, the aim of this study was to characterize the surfaceome of primary equine chondrocytes isolated from healthy joints following exposure to the pro-inflammatory cytokines interleukin-1-beta (IL-1β) and tumour necrosis factor-alpha (TNF-α). We employed combined methodology that we recently developed for investigating the surfaceome in stem cells. Membrane proteins were isolated using an aminooxy-biotinylation technique and analysed by mass spectrometry using high throughput shotgun proteomics. Selected proteins were validated by western blotting. Results Amongst the 431 unique cell surface proteins identified, a high percentage of low-abundance proteins, such as ion channels, receptors and transporter molecules were detected. Data are available via ProteomeXchange with identifier PXD014773. A high number of proteins exhibited different expression patterns following chondrocyte stimulation with pro-inflammatory cytokines. Low density lipoprotein related protein 1 (LPR-1), thrombospondin-1 (TSP-1), voltage dependent anion channel (VDAC) 1–2 and annexin A1 were considered to be of special interest and were analysed further by western blotting. Conclusions Our results provide, for the first time, a repository for proteomic data on differentially expressed low-abundance membrane proteins on the surface of chondrocytes in response to pro-inflammatory stimuli.
Collapse
Affiliation(s)
- Bernadette Jeremiasse
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Christopher R Fellows
- Department of Veterinary Pre-Clinical Sciences, School of Veterinary Science and Medicine, University of Surrey, Guildford, UK
| | - David J Boocock
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | | | | | - Floris Lafeber
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Willem E van Spil
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Ali Mobasheri
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands. .,Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland. .,Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania. .,Centre for Sport, Exercise and Osteoarthritis Research Versus Arthritis, Queen's Medical Centre, Nottingham, UK. .,Department of Orthopedics, UMC Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
27
|
Mannella CA. Consequences of Folding the Mitochondrial Inner Membrane. Front Physiol 2020; 11:536. [PMID: 32581834 PMCID: PMC7295984 DOI: 10.3389/fphys.2020.00536] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/30/2020] [Indexed: 12/18/2022] Open
Abstract
A fundamental first step in the evolution of eukaryotes was infolding of the chemiosmotic membrane of the endosymbiont. This allowed the proto-eukaryote to amplify ATP generation while constraining the volume dedicated to energy production. In mitochondria, folding of the inner membrane has evolved into a highly regulated process that creates specialized compartments (cristae) tuned to optimize function. Internalizing the inner membrane also presents complications in terms of generating the folds and maintaining mitochondrial integrity in response to stresses. This review describes mechanisms that have evolved to regulate inner membrane topology and either preserve or (when appropriate) rupture the outer membrane.
Collapse
Affiliation(s)
- Carmen A. Mannella
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
28
|
Kanwar P, Samtani H, Sanyal SK, Srivastava AK, Suprasanna P, Pandey GK. VDAC and its interacting partners in plant and animal systems: an overview. Crit Rev Biotechnol 2020; 40:715-732. [PMID: 32338074 DOI: 10.1080/07388551.2020.1756214] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Molecular trafficking between different subcellular compartments is the key for normal cellular functioning. Voltage-dependent anion channels (VDACs) are small-sized proteins present in the outer mitochondrial membrane, which mediate molecular trafficking between mitochondria and cytoplasm. The conductivity of VDAC is dependent on the transmembrane voltage, its oligomeric state and membrane lipids. VDAC acts as a convergence point to a diverse variety of mitochondrial functions as well as cell survival. This functional diversity is attained due to their interaction with a plethora of proteins inside the cell. Although, there are hints toward functional conservation/divergence between animals and plants; knowledge about the functional role of the VDACs in plants is still limited. We present here a comparative overview to provide an integrative picture of the interactions of VDAC with different proteins in both animals and plants. Also discussed are their physiological functions from the perspective of cellular movements, signal transduction, cellular fate, disease and development. This in-depth knowledge of the biological importance of VDAC and its interacting partner(s) will assist us to explore their function in the applied context in both plant and animal.
Collapse
Affiliation(s)
- Poonam Kanwar
- Department of Plant Molecular Biology, University of Delhi South Campus, New Delhi, India
| | - Harsha Samtani
- Department of Plant Molecular Biology, University of Delhi South Campus, New Delhi, India
| | - Sibaji K Sanyal
- Department of Plant Molecular Biology, University of Delhi South Campus, New Delhi, India
| | - Ashish K Srivastava
- Nuclear Agriculture and Biotechnology Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Penna Suprasanna
- Nuclear Agriculture and Biotechnology Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Girdhar K Pandey
- Department of Plant Molecular Biology, University of Delhi South Campus, New Delhi, India
| |
Collapse
|
29
|
Malik C, Ghosh S. Quinidine partially blocks mitochondrial voltage-dependent anion channel (VDAC). EUROPEAN BIOPHYSICS JOURNAL: EBJ 2020; 49:193-205. [DOI: 10.1007/s00249-020-01426-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 02/12/2020] [Accepted: 02/17/2020] [Indexed: 02/07/2023]
|
30
|
Xie D, Audi SH, Dash RK. A size-modified poisson-boltzmann ion channel model in a solvent of multiple ionic species: Application to voltage-dependent anion channel. J Comput Chem 2020; 41:218-230. [PMID: 31845398 PMCID: PMC8189662 DOI: 10.1002/jcc.26091] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/30/2019] [Accepted: 09/24/2019] [Indexed: 12/30/2022]
Abstract
We present a new size-modified Poisson-Boltzmann ion channel (SMPBIC) model and use it to calculate the electrostatic potential, ionic concentrations, and electrostatic solvation free energy for a voltage-dependent anion channel (VDAC) on a biological membrane in a solution mixture of multiple ionic species. In particular, the new SMPBIC model adopts a membrane surface charge density and a natural Neumann boundary condition to reflect the charge effect of the membrane on the electrostatics of VDAC. To avoid the singularity difficulties caused by the atomic charges of VDAC, the new SMPBIC model is split into three submodels such that the solution of one of the submodels is obtained analytically and contains all the singularity points of the SMPBIC model. The other two submodels are then solved numerically much more efficiently than the original SMPBIC model. As an application of this SMPBIC submodel partitioning scheme, we derive a new formula for computing the electrostatic solvation free energy. Numerical results for a human VDAC isoform 1 (hVDAC1) in three different salt solutions, each with up to five different ionic species, confirm the significant effects of membrane surface charges on both the electrostatics and ionic concentrations. The results also show that the new SMPBIC model can describe well the anion selectivity property of hVDAC1, and that the new electrostatic solvation free energy formula can significantly improve the accuracy of the currently used formula. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Dexuan Xie
- Department of Mathematical Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, 53201
| | - Said H Audi
- Department of Biomedical Engineering, Marquette University, Milwaukee, Wisconsin, 53233
| | - Ranjan K Dash
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin, 53226
| |
Collapse
|
31
|
Zhu Y, Liu Q, Liao M, Diao L, Wu T, Liao W, Wang Z, Li B, Zhang S, Wang S, Xie W, Jiang Y, Xu N, Zeng Y, Yang BB, Zhang Y. Overexpression of lncRNA EPB41L4A-AS1 Induces Metabolic Reprogramming in Trophoblast Cells and Placenta Tissue of Miscarriage. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:518-532. [PMID: 31671345 PMCID: PMC6838551 DOI: 10.1016/j.omtn.2019.09.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 05/26/2019] [Accepted: 09/14/2019] [Indexed: 11/20/2022]
Abstract
Long non-coding RNAs (lncRNAs) have been shown to be crucial regulators in numerous human diseases. However, little is known about their effects on early recurrent miscarriage (RM). Here we aimed to investigate the role of lncRNA EPB41L4A-AS1 on placental trophoblast cell metabolic reprogramming, which might be involved in the pathogenesis of RM. After microarray and GEO database analyses, we found that EPB41L4A-AS1 was significantly increased in early RM placental tissue, and this increase may relate to estradiol-mediated upregulation of PGC-1α. EPB41L4A-AS1 overexpression inhibits glycolysis but increases the dependence on fatty acid oxidation in mitochondrion metabolism and suppresses the Warburg effect, which is necessary for rapid growth of the placental villus, leading to miscarriage. Mechanistic analyses demonstrated that EPB41L4A-AS1 functions as a lncRNA in the regulation of VDAC1 and HIF-1α expression through enhancement of H3K4me3 levels in the promoters of VDAC1 and HIF1A-AS1, a natural antisense transcript (NAT) lncRNA of HIF-1α. Taken together, these findings demonstrate that aberrant expression of EPB41L4A-AS1 is involved in the etiology of early RM, and it may be a candidate diagnostic hallmark and a potential therapeutic target for early RM treatment.
Collapse
Affiliation(s)
- Yuanchang Zhu
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China; State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China
| | - Qing Liu
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China; State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China
| | - Meijian Liao
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China; State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China
| | - Lianghui Diao
- Shenzhen Key Laboratory for Reproductive Immunology of Preimplantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518055, P.R. China
| | - Tonghua Wu
- Shenzhen Key Laboratory for Reproductive Immunology of Preimplantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518055, P.R. China
| | - Weijie Liao
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China
| | - Ziqiang Wang
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China
| | - Bing Li
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China
| | - Shikuan Zhang
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China
| | - Songmao Wang
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China
| | - Weidong Xie
- Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China; State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China; Open FIESTA Center, Tsinghua University, Shenzhen 518055, P.R. China
| | - Yuyang Jiang
- Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China
| | - Naihan Xu
- Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China; State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China; Open FIESTA Center, Tsinghua University, Shenzhen 518055, P.R. China
| | - Yong Zeng
- Shenzhen Key Laboratory for Reproductive Immunology of Preimplantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518055, P.R. China
| | - Burton B Yang
- Sunnybrook Research Institute and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| | - Yaou Zhang
- Key Lab in Healthy Science and Technology, Division of Life Science, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China; State Key Laboratory of Chemical Oncogenomics, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, P.R. China; Open FIESTA Center, Tsinghua University, Shenzhen 518055, P.R. China.
| |
Collapse
|
32
|
Zhu C, Tian L, Yang H, Chen P, Li Y, Liu Y. Mitochondrial outer membrane voltage-dependent anion channel is involved in renal dysfunction in a spontaneously hypertensive rat carrying transfer RNA mutations. Eur J Pharmacol 2019; 865:172622. [PMID: 31618620 DOI: 10.1016/j.ejphar.2019.172622] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 08/13/2019] [Accepted: 08/20/2019] [Indexed: 11/18/2022]
Abstract
Mitochondrial DNA mutations promote hypertensive renal dysfunction, but the molecular mechanism remains unclear. This study compared renal damage between spontaneously hypertensive rats (SHR) and SHR with mitochondrial transfer (t)RNA mutations. To investigate the role of mitochondrial outer membrane voltage-dependent anion channel 1 (VDAC1) in the process of tRNA-promoting mitochondrial dysfunction, we treated HK-2 cells with H2O2, cyclosporine (CsA), or atractylodin (Atr) to observe the association between VDAC1 and mitochondrial function. Intriguingly, the mitochondrial structure of SHR carrying tRNA mutations was obviously disordered, and reactive oxygen species production and VDAC1 and Bax expression and binding were increased, which was associated with marked renal dysfunction. The expression of VDAC1 and Bax was also up-regulated in HK-2 cells by H2O2 treatment. However, CsA and Atr had no significant effect on the expression of VDAC1 and Bax. H2O2 caused mitochondrial membrane potential collapse, while CsA could increase the mitochondrial membrane potential and Atr had the opposite effect. Treatment with H2O2 significantly decreased ATP synthesis, which was improved by intervention with Atr. H2O2 also decreased the maximum oxygen consumption rate, while CsA and Atr had no significant effect. We found that H2O2 promoted the colocalization of VDAC1 and Bax, which was partially inhibited by intervention with CsA or Atr. In conclusion, we found that tRNA mutations promoted hypertensive renal insufficiency. Increased reactive oxygen species was an important associated mechanism, which inhibited mitochondrial function by affecting VDAC1 expression and function.
Collapse
Affiliation(s)
- Chao Zhu
- Cardiac Department, Institute of Geriatric Cardiology, PLA General Hospital, Beijing, 100853, China; Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Liuyang Tian
- Cardiac Department, Institute of Geriatric Cardiology, PLA General Hospital, Beijing, 100853, China; Department of Cardiology, Tianjin Union Medical Center, Tianjin, 300121, China
| | - Huanwan Yang
- Cardiac Department, Institute of Geriatric Cardiology, PLA General Hospital, Beijing, 100853, China
| | - Pu Chen
- National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yang Li
- Cardiac Department, Institute of Geriatric Cardiology, PLA General Hospital, Beijing, 100853, China.
| | - Yuqi Liu
- Cardiac Department, Institute of Geriatric Cardiology, PLA General Hospital, Beijing, 100853, China; National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, 100853, China; Department of Cardiology & National Clinical Research Center of Geriatric Disease, Chinese PLA General Hospital, Beijing, 100853, China; Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
33
|
Cheng WW, Budelier MM, Sugasawa Y, Bergdoll L, Queralt-Martín M, Rosencrans W, Rostovtseva TK, Chen ZW, Abramson J, Krishnan K, Covey DF, Whitelegge JP, Evers AS. Multiple neurosteroid and cholesterol binding sites in voltage-dependent anion channel-1 determined by photo-affinity labeling. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1269-1279. [PMID: 31176038 PMCID: PMC6681461 DOI: 10.1016/j.bbalip.2019.06.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/23/2019] [Accepted: 06/02/2019] [Indexed: 12/31/2022]
Abstract
Voltage-dependent anion channel-1 (VDAC1) is a mitochondrial porin that is implicated in cellular metabolism and apoptosis, and modulated by numerous small molecules including lipids. VDAC1 binds sterols, including cholesterol and neurosteroids such as allopregnanolone. Biochemical and computational studies suggest that VDAC1 binds multiple cholesterol molecules, but photolabeling studies have identified only a single cholesterol and neurosteroid binding site at E73. To identify all the binding sites of neurosteroids in VDAC1, we apply photo-affinity labeling using two sterol-based photolabeling reagents with complementary photochemistry: 5α-6-AziP which contains an aliphatic diazirine, and KK200 which contains a trifluoromethyl-phenyldiazirine (TPD) group. 5α-6-AziP and KK200 photolabel multiple residues within an E73 pocket confirming the presence of this site and mapping sterol orientation within this pocket. In addition, KK200 photolabels four other sites consistent with the finding that VDAC1 co-purifies with five cholesterol molecules. Both allopregnanolone and cholesterol competitively prevent photolabeling at E73 and three other sites indicating that these are common sterol binding sites shared by both neurosteroids and cholesterol. Binding at the functionally important residue E73 suggests a possible role for sterols in regulating VDAC1 signaling and interaction with partner proteins.
Collapse
Affiliation(s)
- Wayland W.L. Cheng
- Department of Anesthesiology, Washington University in St. Louis, MO 63110, USA
| | - Melissa M. Budelier
- Department of Anesthesiology, Washington University in St. Louis, MO 63110, USA,Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, MO 63110, USA
| | - Yusuke Sugasawa
- Department of Anesthesiology, Washington University in St. Louis, MO 63110, USA
| | - Lucie Bergdoll
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - María Queralt-Martín
- Section on Molecular Transport, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - William Rosencrans
- Section on Molecular Transport, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tatiana K. Rostovtseva
- Section on Molecular Transport, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zi-Wei Chen
- Department of Anesthesiology, Washington University in St. Louis, MO 63110, USA,Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis, MO 63110, USA
| | - Jeff Abramson
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Kathiresan Krishnan
- Department of Developmental Biology, Washington University in St. Louis, MO 63110, USA
| | - Douglas F. Covey
- Department of Anesthesiology, Washington University in St. Louis, MO 63110, USA,Department of Developmental Biology, Washington University in St. Louis, MO 63110, USA,Department of Psychiatry, Washington University in St. Louis, MO 63110, USA,Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis, MO 63110, USA
| | - Julian P. Whitelegge
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Alex S. Evers
- Department of Anesthesiology, Washington University in St. Louis, MO 63110, USA,Department of Developmental Biology, Washington University in St. Louis, MO 63110, USA,Department of Psychiatry, Washington University in St. Louis, MO 63110, USA,Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis, MO 63110, USA,Corresponding author at: Department of Anesthesiology, Washington University School of Medicine, Campus Box 8054, St. Louis, MO 63110, USA. (A.S. Evers)
| |
Collapse
|
34
|
Seo JH, Park HS, Park SS, Kim CJ, Kim DH, Kim TW. Physical exercise ameliorates psychiatric disorders and cognitive dysfunctions by hippocampal mitochondrial function and neuroplasticity in post-traumatic stress disorder. Exp Neurol 2019; 322:113043. [PMID: 31446079 DOI: 10.1016/j.expneurol.2019.113043] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 07/24/2019] [Accepted: 08/20/2019] [Indexed: 12/15/2022]
Abstract
Post-traumatic stress disorder (PTSD) is a stress-related condition that can be triggered by witnessing or experiencing a life-threatening event, such as a war, natural disaster, terrorist attack, major accident, or assault. PTSD is caused by dysfunction of the hippocampus and causes problems associated with brain functioning, such as anxiety, depression, and cognitive impairment. Exercise is known to have a positive effect on brain function, especially in the hippocampus. In this study, we investigated the effect of aerobic exercise on mitochondrial function and neuroplasticity in the hippocampus as well as behavioral changes in animal models of PTSD. Exposure to severe stress resulted in mitochondrial dysfunction in the hippocampus, including impaired Ca2+ homeostasis, an increase in reactive oxygen species such as H2O2, a decrease in the O2 respiration rate, and overexpression of membrane permeability transition pore-related proteins, including voltage-dependent anion channel, adenine nucleotide translocase, and cyclophilin-D. Exposure to extreme stress also decreased neuroplasticity by increasing apoptosis and decreasing the brain-derived neurotrophic factor level and neurogenesis, resulting in increased anxiety, depression, and cognitive impairment. The impairments in mitochondrial function and neuroplasticity in the hippocampus, as well as anxiety, depression, and cognitive impairment, were all improved by exercise. Exercise-induced improvement of the brain-derived neurotrophic factor level in particular might alter mitochondrial function, neuroplasticity, and the rate of apoptosis in the hippocampus. Therefore, exercise might be an important non-pharmacological intervention for the prevention and treatment of the pathobiology of PTSD.
Collapse
Affiliation(s)
- Jin-Hee Seo
- Department of Adapted physical education, Baekseok University, Cheonan, Republic of Korea
| | - Hye-Sang Park
- Department of Kinesiology, College of public health and Cardiovascular Research Center, Lewis Katz school of Medicine, Temple University, Philadelphia, PA, USA
| | - Sang-Seo Park
- Department of physiology, College of medicine, Kyung Hee University, Seoul, Republic of Korea; Kohwang Medical Research Institute, Kyung Hee University, Seoul, Republic of Korea
| | - Chang-Ju Kim
- Department of physiology, College of medicine, Kyung Hee University, Seoul, Republic of Korea; Kohwang Medical Research Institute, Kyung Hee University, Seoul, Republic of Korea
| | - Dong-Hyun Kim
- College of Sports science, Sungkyunkwan University, Suwon, Republic of Korea
| | - Tae-Woon Kim
- Department of physiology, College of medicine, Kyung Hee University, Seoul, Republic of Korea; Kohwang Medical Research Institute, Kyung Hee University, Seoul, Republic of Korea; Exercise Rehabilitation Research Institute, Department of Exercise & Health Science, Sangmyung University, Seoul, Republic of Korea.
| |
Collapse
|
35
|
Abstract
The programmed release of apoptogenic proteins from mitochondria is a core event of apoptosis, although ancestral roles of this phenomenon are not known. In mammals, one such apoptogenic protein is Endonuclease G (EndoG), a conserved mitochondrial nuclease that fragments the DNA of dying cells. In this work, we show that budding yeast executes meiotically programmed mitochondrial release of an EndoG homolog, Nuc1, during sporulation. In contrast to EndoG's ostensible pro-death function during apoptosis, Nuc1 mitochondrial release is pro-survival, attenuating the cytosolic L-A and Killer double-stranded RNA mycoviruses and protecting meiotic progeny from the catastrophic consequences of their derepression. The protective viral attenuation role of this pathway illuminates a primordial role for mitochondrial release of EndoG, and perhaps of apoptosis itself.
Collapse
|
36
|
Zhang L, Zhang J, Ye Z, Manevich Y, Ball LE, Bethard JR, Jiang YL, Broome AM, Dalton AC, Wang GY, Townsend DM, Tew KD. Isoflavone ME-344 Disrupts Redox Homeostasis and Mitochondrial Function by Targeting Heme Oxygenase 1. Cancer Res 2019; 79:4072-4085. [PMID: 31227482 DOI: 10.1158/0008-5472.can-18-3503] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 02/12/2019] [Accepted: 06/17/2019] [Indexed: 12/19/2022]
Abstract
ME-344 is a second-generation isoflavone with unusual cytotoxic properties that is in clinical testing in cancer. To identify targets that contribute to its anticancer activity and therapeutic index, we used lung cancer cell lines that are naturally sensitive or resistant to ME-344. Drug-induced apoptosis was linked with enhanced levels of reactive oxygen species and this initiated a nuclear erythroid factor 2-like 2 signaling response, downstream of which, heme oxygenase 1 (HO-1) was also found to be time-dependently inhibited by ME-344. ME-344 specifically bound to, and altered, HO-1 structure and increased HO-1 translocation from the rough endoplasmic reticulum to mitochondria, but only in drug-sensitive cells. These effects did not occur in either drug-resistant or primary lung fibroblasts with lower HO-1 basal levels. HO-1 was confirmed as a drug target by using surface plasmon resonance technology and through interaction with a clickable ME-344 compound (M2F) and subsequent proteomic analyses, showing direct binding of ME-344 with HO-1. Proteomic analysis showed that clusters of mitochondrial proteins, including voltage-dependent anion-selective channels, were also impacted by ME-344. Human lung cancer biopsies expressed higher levels of Nrf2 and HO-1 compared with normal tissues. Overall, our data show that ME-344 inhibits HO-1 and impacts its mitochondrial translocation. Other mitochondrial proteins are also affected, resulting in interference in tumor cell redox homeostasis and mitochondrial function. These factors contribute to a beneficial therapeutic index and support continued clinical development of ME-344. SIGNIFICANCE: A novel cytotoxic isoflavone is shown to inhibit heme oxygenase, a desirable yet elusive target that disrupts redox homeostasis causing cell death.
Collapse
Affiliation(s)
- Leilei Zhang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina
| | - Jie Zhang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina
| | - Zhiwei Ye
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina
| | - Yefim Manevich
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina
| | - Lauren E Ball
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina
| | - Jennifer R Bethard
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina
| | - Yu-Lin Jiang
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina
| | - Ann-Marie Broome
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina
| | - Annamarie C Dalton
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Gavin Y Wang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Danyelle M Townsend
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina.
| | - Kenneth D Tew
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
37
|
Breda CNDS, Davanzo GG, Basso PJ, Saraiva Câmara NO, Moraes-Vieira PMM. Mitochondria as central hub of the immune system. Redox Biol 2019; 26:101255. [PMID: 31247505 PMCID: PMC6598836 DOI: 10.1016/j.redox.2019.101255] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/01/2019] [Accepted: 06/10/2019] [Indexed: 02/08/2023] Open
Abstract
Nearly 130 years after the first insights into the existence of mitochondria, new rolesassociated with these organelles continue to emerge. As essential hubs that dictate cell fate, mitochondria integrate cell physiology, signaling pathways and metabolism. Thus, recent research has focused on understanding how these multifaceted functions can be used to improve inflammatory responses and prevent cellular dysfunction. Here, we describe the role of mitochondria on the development and function of immune cells, highlighting metabolic aspects and pointing out some metabolic- independent features of mitochondria that sustain cell function.
Collapse
Affiliation(s)
- Cristiane Naffah de Souza Breda
- Transplantation Immunobiology Lab, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Gustavo Gastão Davanzo
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Paulo José Basso
- Transplantation Immunobiology Lab, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Niels Olsen Saraiva Câmara
- Transplantation Immunobiology Lab, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.
| | - Pedro Manoel Mendes Moraes-Vieira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil.
| |
Collapse
|
38
|
Rovini A. Tubulin-VDAC Interaction: Molecular Basis for Mitochondrial Dysfunction in Chemotherapy-Induced Peripheral Neuropathy. Front Physiol 2019; 10:671. [PMID: 31214047 PMCID: PMC6554597 DOI: 10.3389/fphys.2019.00671] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/13/2019] [Indexed: 12/12/2022] Open
Abstract
Tubulin is a well-established target of microtubule-targeting agents (MTAs), a widely used class of chemotherapeutic drugs. Yet, aside from their powerful anti-cancer efficiency, MTAs induce a dose-limiting and debilitating peripheral neurotoxicity. Despite intensive efforts in the development of neuroprotective agents, there are currently no approved therapies to effectively manage chemotherapy-induced peripheral neuropathy (CIPN). Over the last decade, attempts to unravel the pathomechanisms underlying the development of CIPN led to the observation that mitochondrial dysfunctions stand as a common feature associated with axonal degeneration. Concomitantly, mitochondria emerged as crucial players in the anti-cancer efficiency of MTAs. The findings that free dimeric tubulin could be associated with mitochondrial membranes and interact directly with the voltage-dependent anion channels (VDACs) located in the mitochondrial outer membrane strongly suggested the existence of an interplay between both subcellular compartments. The biological relevance of the interaction between tubulin and VDAC came from subsequent in vitro studies, which found dimeric tubulin to be a potent modulator of VDAC and ultimately of mitochondrial membrane permeability to respiratory substrates. Therefore, one of the hypothetic mechanisms of CIPN implies that MTAs, by binding directly to the tubulin associated with VDAC, interferes with mitochondrial function in the peripheral nervous system. We review here the foundations of this hypothesis and discuss them in light of the current knowledge. A focus is set on the molecular mechanisms behind MTA interference with dimeric tubulin and VDAC interaction, the potential relevance of tubulin isotypes and availability as a free dimer in the specific context of MTA-induced CIPN. We further highlight the emerging interest for VDAC and its interacting partners as a promising therapeutic target in neurodegeneration.
Collapse
Affiliation(s)
- Amandine Rovini
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
39
|
Gu Z, Song W, Liu S, Li B, Plant LD, Meng XY. Potential blockade of the human voltage-dependent anion channel by MoS 2 nanoflakes. Phys Chem Chem Phys 2019; 21:9520-9530. [PMID: 31020281 DOI: 10.1039/c9cp00195f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2024]
Abstract
Despite significant interest in molybdenum disulfide (MoS2) nanomaterials, particularly in biomedicine, their biological effects have been understudied. Here, we explored the effect of MoS2 nanoflakes on the ubiquitous mitochondrial porin voltage-dependent anion channel (VDAC1), using a combined computational and functional approach. All-atomic molecular dynamics simulations suggest that MoS2 nanoflakes make specific contact interactions with human VDAC1. We show that the initial contacts between hVDAC1 and the nanoflake are hydrophobic but are subsequently enhanced by a complex interplay of van der Waals (vdW), hydrophobic and electrostatic interactions in the equilibrium state. Moreover, the MoS2 nanoflake can insert into the lumen of the hVDAC1 pore. Free-energy calculations computed by the potential of mean force (PMF) verify that the blocked configuration of the MoS2-hVDAC1 complex is more energetically favorable than the non-blocked binding mode. Consistent with these predictions, we showed that MoS2 depolarizes the mitochondrial membrane potential (Ψm) and causes a decrease in the viability of mammalian tissue culture cells. These findings might shed new light on the potential biological effect of MoS2 nanomaterials.
Collapse
Affiliation(s)
- Zonglin Gu
- Institute of Quantitative Biology and Medicine, SRMP and RAD-X, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | | | | | | | | | | |
Collapse
|
40
|
Voltage-dependent anion channel isoform 3 as a potential male contraceptive drug target. Future Med Chem 2019; 11:857-867. [PMID: 30998114 DOI: 10.4155/fmc-2018-0328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Voltage-dependent anion channel isoform 3 (VDAC3), a channel in the mitochondrial outer membrane, has been suggested to play a role in the regulation of ATP transport and Ca2+ homeostasis. These processes are regarded as important for spermatozoa motility. Accordingly, in previous years, mutations in the VDAC3-encoding gene were detected in spermatozoa with low motility from infertile patients. Therefore, it can be assumed that these mutations would cause alteration of the structure and/or charge of the VDAC3 channel. The review is focused on current knowledge about contribution of VDAC3 activity to human spermatozoa motility and morphology. We also discuss the possibility of designing new molecules that could specifically block the VDAC3 channel and consequently act as male contraceptives.
Collapse
|
41
|
Mokrousov IS, Perfilova VN, Prokofiev II, Popova TA, Vodopyanova EG, Vasil'eva OS, Tyurenkov IN. Effect of a new cyclic derivative of GABA, RGPU-207, on the functions of cardiac and cerebral mitochondria of stressed animals. J Pharm Pharmacol 2019; 71:1055-1064. [DOI: 10.1111/jphp.13086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 02/03/2019] [Indexed: 12/19/2022]
Abstract
Abstract
Objectives
The objective of this study was to investigate the effects of a new derivative of GABA, RGPU-207 compound, on the mitochondrial functions of stressed animals.
Methods
RGPU-207 and the comparator drugs (phenibut and piracetam) were administered intraperitoneally to unstressed and stressed male rats at a dose of 9.4, 25 and 400 mg/kg, respectively. The oxygen consumption by cardiac and cerebral mitochondria in state 3 and 4 and Chance's respiratory control ratio (RCR) was investigated. The concentration of lipid peroxidation products (LPO) such as malondialdehyde (MDA), conjugated dienes (CD) and diketones was evaluated in the isolated mitochondria, as well as the activity of the antioxidant system (AOS) enzymes (superoxide dismutase (SOD), glutathione peroxidase (GP) and catalase).
Key findings
A new cyclic GABA derivative, RGPU-207 compound, at the dose of 9.4 mg/kg promotes a decline in MDA, diketone and CD concentrations in mitochondria and increases the levels of SOD, GP and catalase activity. Mitochondrial functional activity increases: oxygen consumption by cerebral mitochondria in state 4 decreases when complex I of the respiratory chain is activated, while malate-dependent state 3 respiration of cardiac mitochondria tends to increase. RCR of cardiac mitochondria increases when complexes I and II are involved. In cerebral mitochondria, malate-dependent and succinate-dependent RCR rise.
Conclusions
Twenty-four-hour immobilization and pain stress activate LPO processes inhibit the activity of the aos enzymes and decrease the functional activity of cardiac and cerebral mitochondria. RGPU-207 restricts LPO, enhances the antioxidant enzyme activity and improves the mitochondrial respiration. The efficacy of RGPU-207 is comparable with phenibut and piracetam.
Collapse
Affiliation(s)
- Ivan Sergeevich Mokrousov
- Department for Pharmacology and Biopharmacy, Volgograd State Medical University, Volgograd, Russian Federation
| | | | - Igor Igorevich Prokofiev
- Department for Pharmacology and Biopharmacy, Volgograd State Medical University, Volgograd, Russian Federation
| | - Tamara Alexandrovna Popova
- Department for Basic and Clinical Biochemistry, Volgograd State Medical University, Volgograd, Russian Federation
| | | | - Olga Sergeevna Vasil'eva
- Department for Organic Chemistry, Herzen State Pedagogical University, St.Petersburg, Russian Federation
| | - Ivan Nikolaevich Tyurenkov
- Department for Pharmacology and Biopharmacy, Volgograd State Medical University, Volgograd, Russian Federation
| |
Collapse
|
42
|
ROS Induced by KillerRed Targeting Mitochondria (mtKR) Enhances Apoptosis Caused by Radiation via Cyt c/Caspase-3 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4528616. [PMID: 30984335 PMCID: PMC6431512 DOI: 10.1155/2019/4528616] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/02/2019] [Accepted: 01/22/2019] [Indexed: 12/12/2022]
Abstract
During radiotherapy, reactive oxygen species- (ROS-) induced apoptosis is one of the main mechanism of radiation. Based on KillerRed which can induce ROS burst in different cell substructures, here we hypothesized that KillerRed targeting mitochondria (mtKR) could induce ROS to enhance apoptosis by radiation. In this study, empty vector, mtKR, and mtmCherry plasmids were successfully constructed, and mitochondrial localization were detected in COS-7 and HeLa cells. After HeLa cells were transfected and irradiated by visible light and X-rays, ROS levels, mitochondrial membrane potential (Δψm), ATPase activities, adenosine triphosphate (ATP) content, apoptosis, and the expressions of mRNA and protein were measured, respectively. Data demonstrated that the ROS levels significantly increased after light exposure, and adding extra radiation, voltage-dependent anion channel 1 (VDAC1) protein increased in the mitochondria, while Na+-K+ and Ca2+-Mg2+ ATPase activities, ATP content, and Δψm significantly reduced. Additionally, the cell apoptotic rates dramatically increased, which referred to the increase of cytochrome c (Cyt c), caspase-9, and caspase-3 mRNA expressions, and Cyt c protein was released from the mitochondria into the cytoplasm; caspase-9 and -3 were activated. These results indicated that mtKR can increase the production of ROS, enhance mitochondrial dysfunction, and strengthen apoptosis by radiation via Cyt c/caspase-3 pathway.
Collapse
|
43
|
Penkauskas T, Preta G. Biological applications of tethered bilayer lipid membranes. Biochimie 2019; 157:131-141. [DOI: 10.1016/j.biochi.2018.11.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/19/2018] [Indexed: 12/16/2022]
|
44
|
Queralt-Martín M, Bergdoll L, Jacobs D, Bezrukov SM, Abramson J, Rostovtseva TK. Assessing the role of residue E73 and lipid headgroup charge in VDAC1 voltage gating. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2019; 1860:22-29. [PMID: 30412693 PMCID: PMC8283775 DOI: 10.1016/j.bbabio.2018.11.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 10/05/2018] [Accepted: 11/04/2018] [Indexed: 12/25/2022]
Abstract
The voltage-dependent anion channel (VDAC) is the most abundant protein of the mitochondrial outer membrane (MOM) where it regulates transport of ions and metabolites in and out of the organelle. VDAC function is extensively studied in a lipid bilayer system that allows conductance monitoring of reconstituted channels under applied voltage. The process of switching from a high-conductance state, open to metabolites, to a variety of low-conducting states, which excludes metabolite transport, is termed voltage gating and the mechanism remains poorly understood. Recent studies have implicated the involvement of the membrane-solvated residue E73 in the gating process through β-barrel destabilization. However, there has been no direct experimental evidence of E73 involvement in VDAC1 voltage gating. Here, using electrophysiology measurements, we exclude the involvement of E73 in murine VDAC1 (mVDAC1) voltage gating process. With an established protocol of assessing voltage gating of VDACs reconstituted into planar lipid membranes, we definitively show that mVDAC1 gating properties do not change when E73 is replaced by either a glutamine or an alanine. We further demonstrate that cholesterol has no effect on mVDAC1 gating characteristics, though it was shown that E73 is coordinating residue in the cholesterol binding site. In contrast, we found a pronounced gating effect based on the charge of the phospholipid headgroup, where the positive charge stimulates and negative charge suppresses gating. These findings call for critical evaluation of the existing models of VDAC gating and contribute to our understanding of VDAC's role in control of MOM permeability and regulation of mitochondrial respiration and metabolism.
Collapse
Affiliation(s)
- María Queralt-Martín
- Section on Molecular Transport, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Lucie Bergdoll
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA.
| | - Daniel Jacobs
- Section on Molecular Transport, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sergey M. Bezrukov
- Section on Molecular Transport, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jeff Abramson
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA.
| | - Tatiana K. Rostovtseva
- Section on Molecular Transport, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA,To whom correspondence should be addressed: Tatiana K. Rostovtseva, Section on Molecular Transport, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 9000 Rockville Pike, Bldg. 9, Room 1E-106, Bethesda, MD 20892-0924. Phone: (301) 402-4702, ; Jeff Abramson, Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095. Phone: (310) 825-3913,
| |
Collapse
|
45
|
Nagakannan P, Islam MI, Karimi-Abdolrezaee S, Eftekharpour E. Inhibition of VDAC1 Protects Against Glutamate-Induced Oxytosis and Mitochondrial Fragmentation in Hippocampal HT22 Cells. Cell Mol Neurobiol 2019; 39:73-85. [PMID: 30421242 DOI: 10.1007/s10571-018-0634-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/07/2018] [Indexed: 01/05/2023]
Abstract
The involvement of glutamate in neuronal cell death in neurodegenerative diseases and neurotrauma is mediated through excitotoxicity or oxytosis. The latter process induces oxidative stress via glutamate-mediated inhibition of cysteine transporter xCT, leading to depletion of the cellular glutathione pool. Mitochondrial damage, loss of mitochondrial membrane potential (MMP), and depletion of energy metabolites have been shown in this process. The Voltage-Dependent Anion Channel-1 (VDAC1) is one of the main components of the mitochondrial outer membrane and plays a gatekeeping role in mitochondria-cytoplasm transport of metabolites. In this study, we explored the possible participation of VDAC-1 in the pathophysiology of oxytosis. Administration of glutamate in HT22 cells that lack the glutamate ionotropic receptors induced an upregulation and oligomerization of VDAC1. This was associated with an increase in ROS and loss of cell survival. Glutamate-mediated oxytosis in this model also decreased MMP and promoted ATP depletion, resulting in translocation of cytochrome c (cyt C) and apoptosis inducing factor (AIF) from mitochondria into the cytosol. This was also accompanied by cleavage of AIF to form truncated AIF. Inhibition of VDAC1 oligomerization using 4,4'-Diisothiocyanatostilbene-2,2'-disulfonate (DIDS), significantly improved the cell survival, decreased the ROS levels, improved mitochondrial functions, and decreased the mitochondrial damage. Notably, DIDS also inhibited the mitochondrial fragmentation caused by glutamate, indicating the active role of VDAC1 oligomerization in the process of mitochondrial fragmentation in oxytosis. These results suggest a critical role for VDAC1 in mitochondrial fragmentation and its potential therapeutic value against glutamate-mediated oxidative neurotoxicity.
Collapse
Affiliation(s)
- Pandian Nagakannan
- Department of Physiology and Pathophysiology, Regenerative Medicine Program and Spinal Cord Research Centre, University of Manitoba, Winnipeg, Canada
| | - Md Imamul Islam
- Department of Physiology and Pathophysiology, Regenerative Medicine Program and Spinal Cord Research Centre, University of Manitoba, Winnipeg, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program and Spinal Cord Research Centre, University of Manitoba, Winnipeg, Canada
| | - Eftekhar Eftekharpour
- Department of Physiology and Pathophysiology, Regenerative Medicine Program and Spinal Cord Research Centre, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
46
|
Zhang H, Feng YW, Yao YM. Potential therapy strategy: targeting mitochondrial dysfunction in sepsis. Mil Med Res 2018; 5:41. [PMID: 30474573 PMCID: PMC6260865 DOI: 10.1186/s40779-018-0187-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 11/08/2018] [Indexed: 12/21/2022] Open
Abstract
Recently, the definition of sepsis was concluded to be a life-threatening organ dysfunction caused by a dysregulated host response to infection. Severe patients always present with uncorrectable hypotension or hyperlactacidemia, which is defined as septic shock. The new definition emphasizes dysregulation of the host response and multiple organ dysfunction, which is partially attributed to metabolic disorders induced by energy crisis and oxidative stress. Mitochondria are a cellular organelle that are well known as the center of energy production, and mitochondrial damage or dysfunction is commonly induced in septic settings and is a predominant factor leading to a worse prognosis. In the present review, we determine the major mitochondrial disorders from morphology to functions in sepsis. In the following, several clinical or pre-clinical assays for monitoring mitochondrial function are demonstrated according to accumulated evidence, which is the first step of specific therapy targeting to modulate mitochondrial function. Accordingly, various reagents used for regulating mitochondrial enzyme activities and promoting biogenesis have been documented, among which mitochondria-targeted cation, TPP-conjugated antioxidants are the most valuable for future trials and clinical treatment to improve mitochondrial function as they may take advantage of the prognosis associated with septic complications.
Collapse
Affiliation(s)
- Hui Zhang
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Fucheng Road 51, Haidian District, Beijing, 100048, China
| | - Yong-Wen Feng
- Department of Critical Care Medicine, The Second People's Hospital of Shenzhen, Shenzhen, 518035, China
| | - Yong-Ming Yao
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Fucheng Road 51, Haidian District, Beijing, 100048, China.
| |
Collapse
|
47
|
Yang M, Xu Y, Heisner JS, Sun J, Stowe DF, Kwok WM, Camara AKS. Peroxynitrite nitrates adenine nucleotide translocase and voltage-dependent anion channel 1 and alters their interactions and association with hexokinase II in mitochondria. Mitochondrion 2018; 46:380-392. [PMID: 30391711 DOI: 10.1016/j.mito.2018.10.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/26/2018] [Accepted: 10/22/2018] [Indexed: 12/17/2022]
Abstract
Cardiac ischemia and reperfusion (IR) injury induces excessive emission of deleterious reactive O2 and N2 species (ROS/RNS), including the non-radical oxidant peroxynitrite (ONOO-) that can cause mitochondria dysfunction and cell death. In this study, we explored whether IR injury in isolated hearts induces tyrosine nitration of adenine nucleotide translocase (ANT) and alters its interaction with the voltage-dependent anion channel 1 (VDAC1). We found that IR injury induced tyrosine nitration of ANT and that exposure of isolated cardiac mitochondria to ONOO- induced ANT tyrosine, Y81, nitration. The exposure of isolated cardiac mitochondria to ONOO- also led ANT to form high molecular weight proteins and dissociation of ANT from VDAC1. We found that IR injury in isolated hearts, hypoxic injury in H9c2 cells, and ONOO- treatment of H9c2 cells and isolated mitochondria, each decreased mitochondrial bound-hexokinase II (HK II), which suggests that ONOO- caused HK II to dissociate from mitochondria. Moreover, we found that mitochondria exposed to ONOO- induced VDAC1 oligomerization which may decrease its binding with HK II. We have reported that ONOO- produced during cardiac IR injury induced tyrosine nitration of VDAC1, which resulted in conformational changes of the protein and increased channel conductance associated with compromised cardiac function on reperfusion. Thus, our results imply that ONOO- produced during IR injury and hypoxic stress impeded HK II association with VDAC1. ONOO- exposure nitrated mitochondrial proteins and also led to cytochrome c (cyt c) release from mitochondria. In addition, in isolated mitochondria exposed to ONOO- or obtained after IR, there was significant compromise in mitochondrial respiration and delayed repolarization of membrane potential during oxidative (ADP) phosphorylation. Taken together, ONOO- produced during cardiac IR injury can nitrate tyrosine residues of two key mitochondrial membrane proteins involved in bioenergetics and energy transfer to contribute to mitochondrial and cellular dysfunction.
Collapse
Affiliation(s)
- Meiying Yang
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yanji Xu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Preventive Medicine, Medical College of Yanbian University, Yanji, Jilin, China
| | - James S Heisner
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jie Sun
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA; Institute of Clinical Medicine Research, Suzhou Hospital affiliated with Nanjing Medical University, Suzhou, Jiangsu, China; Department of Gastroenterology and Hepatology, Suzhou Hospital affiliated with Nanjing Medical University, Suzhou, Jiangsu, China
| | - David F Stowe
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, WI, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA; Research Service, Zablocki VA Medical Center, Milwaukee, WI, USA
| | - Wai-Meng Kwok
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA; Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Amadou K S Camara
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA; Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
48
|
Kamei Y, Koushi M, Aoyama Y, Asakai R. The yeast mitochondrial permeability transition is regulated by reactive oxygen species, endogenous Ca 2+ and Cpr3, mediating cell death. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2018; 1859:1313-1326. [PMID: 30031690 DOI: 10.1016/j.bbabio.2018.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 11/29/2022]
Abstract
We investigated the properties of the permeability transition pore (PTP) in Saccharomyces cerevisiae in agar-embedded mitochondria (AEM) and agar-embedded cells (AEC) and its role in yeast death. In AEM, ethanol-induced pore opening, as indicated by the release of calcein and mitochondrial membrane depolarization, can be inhibited by CsA, by Cpr3 deficiency, and by the antioxidant glutathione. Notably, the pore opening is inhibited, when mitochondria are preloaded by EGTA or Fluo3 to chelate matrix Ca2+, or are pretreated with 4-Br A23187 to extract matrix Ca2+, prior to agar-embedding, or when pore opening is induced in the presence of EGTA; opened pores are re-closed by sequential treatment with CsA, 4-Br A23187 plus EGTA and NADH, indicating endogenous matrix Ca2+ involvement. CsA also inhibits the pore opening with low conductance triggered by exogenous Ca2+ transport with ETH129. In AEC, the treatment of tert-butylhydroperoxide, a pro-oxidant that triggers transient pore opening in high conductance in AEM, induces yeast death, which is also dependent on CsA and Cpr3. Furthermore, AEMs from mutants lacking three ADP/ATP carrier (AAC) isoforms and with defective ATP synthase dimerization exhibit high and low conductance pore openings with CsA sensitivity, respectively. Collectively, these data show that the yeast PTP is regulated by Cpr3, endogenous matrix Ca2+, and reactive oxygen species, and that it is involved in yeast death; furthermore, ATP synthase dimers play a key role in CsA-sensitive pore formation, while AACs are dispensable.
Collapse
Affiliation(s)
- Yoshiko Kamei
- Department of Morphophysiology, Faculty of Pharmaceutical Sciences, Josai International University, 1 Gumyo, Togane, Chiba 283-8555, Japan
| | - Masami Koushi
- Department of Morphophysiology, Faculty of Pharmaceutical Sciences, Josai International University, 1 Gumyo, Togane, Chiba 283-8555, Japan
| | - Yasunori Aoyama
- Department of Morphophysiology, Faculty of Pharmaceutical Sciences, Josai International University, 1 Gumyo, Togane, Chiba 283-8555, Japan
| | - Rei Asakai
- Department of Morphophysiology, Faculty of Pharmaceutical Sciences, Josai International University, 1 Gumyo, Togane, Chiba 283-8555, Japan.
| |
Collapse
|
49
|
Moles A, Torres S, Baulies A, Garcia-Ruiz C, Fernandez-Checa JC. Mitochondrial-Lysosomal Axis in Acetaminophen Hepatotoxicity. Front Pharmacol 2018; 9:453. [PMID: 29867464 PMCID: PMC5968389 DOI: 10.3389/fphar.2018.00453] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/18/2018] [Indexed: 12/19/2022] Open
Abstract
Acetaminophen (APAP) toxicity is the most common cause of acute liver failure and a major indication for liver transplantion in the United States and Europe. Although significant progress has been made in understanding the molecular mechanisms underlying APAP hepatotoxicity, there is still an urgent need to find novel and effective therapies against APAP-induced acute liver failure. Hepatic APAP metabolism results in the production of the reactive metabolite N-acetyl-p-benzoquinone imine (NAPQI), which under physiological conditions is cleared by its conjugation with glutathione (GSH) to prevent its targeting to mitochondria. APAP overdose or GSH limitation leads to mitochondrial NAPQI-protein adducts formation, resulting in oxidative stress, mitochondrial dysfunction, and necrotic cell death. As mitochondria are a major target of APAP hepatotoxicity, mitochondrial quality control and clearance of dysfunctional mitochondria through mitophagy, emerges as an important strategy to limit oxidative stress and the engagement of molecular events leading to cell death. Recent evidence has indicated a lysosomal-mitochondrial cross-talk that regulates APAP hepatotoxicity. Moreover, as lysosomal function is essential for mitophagy, impairment in the fusion of lysosomes with autophagosomes-containing mitochondria may compromise the clearance of dysfunctional mitochondria, resulting in exacerbated APAP hepatotoxicity. This review centers on the role of mitochondria in APAP hepatotoxicity and how the mitochondrial/lysosomal axis can influence APAP-induced liver failure.
Collapse
Affiliation(s)
- Anna Moles
- Cell Death and Proliferation, Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain.,Liver Unit, Clinical and Provincial Hospital of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer and CIBEREHD, Barcelona, Spain
| | - Sandra Torres
- Cell Death and Proliferation, Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain.,Liver Unit, Clinical and Provincial Hospital of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer and CIBEREHD, Barcelona, Spain
| | - Anna Baulies
- Cell Death and Proliferation, Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain.,Liver Unit, Clinical and Provincial Hospital of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer and CIBEREHD, Barcelona, Spain
| | - Carmen Garcia-Ruiz
- Cell Death and Proliferation, Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain.,Liver Unit, Clinical and Provincial Hospital of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer and CIBEREHD, Barcelona, Spain.,Research Center for Alcoholic Liver and Pancreatic Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Jose C Fernandez-Checa
- Cell Death and Proliferation, Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain.,Liver Unit, Clinical and Provincial Hospital of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer and CIBEREHD, Barcelona, Spain.,Research Center for Alcoholic Liver and Pancreatic Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
50
|
Wang X, Niu J, Li J, Shen X, Shen S, Straubinger RM, Qu J. Temporal Effects of Combined Birinapant and Paclitaxel on Pancreatic Cancer Cells Investigated via Large-Scale, Ion-Current-Based Quantitative Proteomics (IonStar). Mol Cell Proteomics 2018; 17:655-671. [PMID: 29358341 PMCID: PMC5880105 DOI: 10.1074/mcp.ra117.000519] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Indexed: 01/05/2023] Open
Abstract
Despite decades of effort, pancreatic adenocarcinoma (PDAC) remains an intractable clinical challenge. An insufficient understanding of mechanisms underlying tumor cell responses to chemotherapy contributes significantly to the lack of effective treatment regimens. Here, paclitaxel, a first-line chemotherapeutic agent, was observed to interact synergistically with birinapant, a second mitochondrial-derived activator of caspases mimetic. Therefore, we investigated molecular-level drug interaction mechanisms using comprehensive, reproducible, and well-controlled ion-current-based MS1 quantification (IonStar). By analyzing 40 biological samples in a single batch, we compared temporal proteomic responses of PDAC cells treated with birinapant and paclitaxel, alone and combined. Using stringent criteria (e.g. strict false-discovery-rate (FDR) control, two peptides/protein), we quantified 4069 unique proteins confidently (99.8% without any missing data), and 541 proteins were significantly altered in the three treatment groups, with an FDR of <1%. Interestingly, most of these proteins were altered only by combined birinapant/paclitaxel, and these predominantly represented three biological processes: mitochondrial function, cell growth and apoptosis, and cell cycle arrest. Proteins responsible for activation of oxidative phosphorylation, fatty acid β-oxidation, and inactivation of aerobic glycolysis were altered largely by combined birinapant/paclitaxel compared with single drugs, suggesting the Warburg effect, which is critical for survival and proliferation of cancer cells, was alleviated by the combination treatment. Metabolic profiling was performed to confirm substantially greater suppression of the Warburg effect by the combined agents compared with either drug alone. Immunoassays confirmed proteomic data revealing changes in apoptosis/survival signaling pathways, such as inhibition of PI3K/AKT, JAK/STAT, and MAPK/ERK signal transduction, as well as induction of G2/M arrest, and showed the drug combination induced much more apoptosis than did single agents. Overall, this in-depth, large-scale proteomics study provided novel insights into molecular mechanisms underlying synergy of combined birinapant/paclitaxel and describes a proteomics/informatics pipeline that can be applied broadly to the development of cancer drug combination regimens.
Collapse
Affiliation(s)
- Xue Wang
- From the ‡Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York 14263
- §New York State Center of Excellence in Bioinformatics and Life Sciences, New York 14203
| | - Jin Niu
- ¶Department of Pharmaceutical Sciences
| | - Jun Li
- §New York State Center of Excellence in Bioinformatics and Life Sciences, New York 14203
| | - Xiaomeng Shen
- §New York State Center of Excellence in Bioinformatics and Life Sciences, New York 14203
- ‖Department of Biochemistry, University at Buffalo, State University of New York, Buffalo, New York 14214
| | - Shichen Shen
- §New York State Center of Excellence in Bioinformatics and Life Sciences, New York 14203
- ‖Department of Biochemistry, University at Buffalo, State University of New York, Buffalo, New York 14214
| | - Robert M Straubinger
- From the ‡Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York 14263;
- §New York State Center of Excellence in Bioinformatics and Life Sciences, New York 14203
- ¶Department of Pharmaceutical Sciences
| | - Jun Qu
- From the ‡Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, New York 14263;
- §New York State Center of Excellence in Bioinformatics and Life Sciences, New York 14203
- ¶Department of Pharmaceutical Sciences
| |
Collapse
|