1
|
Miyauchi A, Watanabe C, Yamada N, Jimbo EF, Kobayashi M, Ohishi N, Nagayoshi A, Aoki S, Kishita Y, Ohtake A, Ohno N, Takahashi M, Yamagata T, Osaka H. Apomorphine is a potent inhibitor of ferroptosis independent of dopaminergic receptors. Sci Rep 2024; 14:4820. [PMID: 38413694 PMCID: PMC10899610 DOI: 10.1038/s41598-024-55293-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 02/22/2024] [Indexed: 02/29/2024] Open
Abstract
Originally, apomorphine was a broad-spectrum dopamine agonist with an affinity for all subtypes of the Dopamine D1 receptor to the D5 receptor. We previously identified apomorphine as a potential therapeutic agent for mitochondrial diseases by screening a chemical library of fibroblasts from patients with mitochondrial diseases. In this study, we showed that apomorphine prevented ferroptosis in fibroblasts from various types of mitochondrial diseases as well as in normal controls. Well-known biomarkers of ferroptosis include protein markers such as prostaglandin endoperoxide synthase 2 (PTGS2), a key gene for ferroptosis-related inflammation PTGS2, lipid peroxidation, and reactive oxygen species. Our findings that apomorphine induced significant downregulation of PTSG2 and suppressed lipid peroxide to the same extent as other inhibitors of ferroptosis also indicate that apomorphine suppresses ferroptosis. To our knowledge, this is the first study to report that the anti-ferroptosis effect of apomorphine is not related to dopamine receptor agonist action and that apomorphine is a potent inhibitor of ferroptotic cell death independent of dopaminergic receptors.
Collapse
Affiliation(s)
- Akihiko Miyauchi
- Department of Pediatrics, Jichi Medical University, Shimotsuke, Japan.
| | - Chika Watanabe
- Department of Pediatrics, Jichi Medical University, Shimotsuke, Japan
| | - Naoya Yamada
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Eriko F Jimbo
- Department of Pediatrics, Jichi Medical University, Shimotsuke, Japan
| | - Mizuki Kobayashi
- Department of Pediatrics, Jichi Medical University, Shimotsuke, Japan
| | - Natsumi Ohishi
- Department of Pediatrics, Jichi Medical University, Shimotsuke, Japan
| | - Atsuko Nagayoshi
- Department of Pediatrics, Jichi Medical University, Shimotsuke, Japan
| | - Shiho Aoki
- Department of Pediatrics, Jichi Medical University, Shimotsuke, Japan
| | - Yoshihito Kishita
- Diagnostics and Therapeutics of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Department of Life Science, Faculty of Science and Engineering, Kindai University, Osaka, Japan
| | - Akira Ohtake
- Department of Clinical Genomics & Pediatrics (Faculty of Medicine), Saitama Medical University, Saitama, Japan
- Center for Intractable Diseases, Saitama Medical University Hospital, Saitama, Japan
| | - Nobuhiko Ohno
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan
- Division of Ultrastructural Research, National Institute for Physiological Sciences, Okazaki, Japan
| | - Masafumi Takahashi
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Takanori Yamagata
- Department of Pediatrics, Jichi Medical University, Shimotsuke, Japan
| | - Hitoshi Osaka
- Department of Pediatrics, Jichi Medical University, Shimotsuke, Japan.
| |
Collapse
|
2
|
Rai SN, Tiwari N, Singh P, Mishra D, Singh AK, Hooshmandi E, Vamanu E, Singh MP. Therapeutic Potential of Vital Transcription Factors in Alzheimer's and Parkinson's Disease With Particular Emphasis on Transcription Factor EB Mediated Autophagy. Front Neurosci 2022; 15:777347. [PMID: 34970114 PMCID: PMC8712758 DOI: 10.3389/fnins.2021.777347] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
Autophagy is an important cellular self-digestion and recycling pathway that helps in maintaining cellular homeostasis. Dysregulation at various steps of the autophagic and endolysosomal pathway has been reported in several neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington disease (HD) and is cited as a critically important feature for central nervous system (CNS) proteostasis. Recently, another molecular target, namely transcription factor EB (TFEB) has been explored globally to treat neurodegenerative disorders. This TFEB, is a key regulator of autophagy and lysosomal biogenesis pathway. Multiple research studies suggested therapeutic potential by targeting TFEB to treat human diseases involving autophagy-lysosomal dysfunction, especially neurodegenerative disorders. A common observation involving all neurodegenerative disorders is their poor efficacy in clearing and recycle toxic aggregated proteins and damaged cellular organelles due to impairment in the autophagy pathway. This dysfunction in autophagy characterized by the accumulation of toxic protein aggregates leads to a progressive loss in structural integrity/functionality of neurons and may even result in neuronal death. In recent years TFEB, a key regulator of autophagy and lysosomal biogenesis, has received considerable attention. It has emerged as a potential therapeutic target in numerous neurodegenerative disorders like AD and PD. In various neurobiology studies involving animal models, TFEB has been found to ameliorate neurotoxicity and rescue neurodegeneration. Since TFEB is a master transcriptional regulator of autophagy and lysosomal biogenesis pathway and plays a crucial role in defining autophagy activation. Studies have been done to understand the mechanisms for TFEB dysfunction, which may yield insights into how TFEB might be targeted and used for the therapeutic strategy to develop a treatment process with extensive application to neurodegenerative disorders. In this review, we explore the role of different transcription factor-based targeted therapy by some natural compounds for AD and PD with special emphasis on TFEB.
Collapse
Affiliation(s)
| | - Neeraj Tiwari
- Faculty of Biosciences, Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Barabanki, India
| | - Payal Singh
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi, India
| | - Divya Mishra
- Centre of Bioinformatics, University of Allahabad, Prayagraj, India
| | - Anurag Kumar Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Etrat Hooshmandi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Emanuel Vamanu
- Faculty of Biotechnology, University of Agronomic Science and Veterinary Medicine, Bucharest, Romania
| | - Mohan P Singh
- Centre of Biotechnology, University of Allahabad, Prayagraj, India
| |
Collapse
|
3
|
Amaldoss MJN, Mehmood R, Yang J, Koshy P, Kumar N, Unnikrishnan A, Sorrell CC. Anticancer Therapeutic Effects of Cerium Oxide Nanoparticles: Known and Unknown Molecular Mechanisms. Biomater Sci 2022; 10:3671-3694. [DOI: 10.1039/d2bm00334a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cerium-based nanoparticles (CeNPs), particularly cerium oxide (CeO2), have been studied extensively for their antioxidant and prooxidant properties. However, their complete redox and enzyme-mimetic mechanisms of therapeutic action at the molecular...
Collapse
|
4
|
Cho HY, Mavi A, Chueng STD, Pongkulapa T, Pasquale N, Rabie H, Han J, Kim JH, Kim TH, Choi JW, Lee KB. Tumor Homing Reactive Oxygen Species Nanoparticle for Enhanced Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:23909-23918. [PMID: 31252451 DOI: 10.1021/acsami.9b07483] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Multifunctional nanoparticles that carry chemotherapeutic agents can be innovative anticancer therapeutic options owing to their tumor-targeting ability and high drug-loading capacity. However, the nonspecific release of toxic DNA-intercalating anticancer drugs from the nanoparticles has significant side effects on healthy cells surrounding the tumors. Herein, we report a tumor homing reactive oxygen species nanoparticle (THoR-NP) platform that is highly effective and selective for ablating malignant tumors. Sodium nitroprusside (SNP) and diethyldithiocarbamate (DDC) were selected as an exogenous reactive oxygen species (ROS) generator and a superoxide dismutase 1 inhibitor, respectively. DDC-loaded THoR-NP, in combination with SNP treatment, eliminated multiple cancer cell lines effectively by the generation of peroxynitrite in the cells (>95% cell death), as compared to control drug treatments of the same concentration of DDC or SNP alone (0% cell death). Moreover, the magnetic core (ZnFe2O4) of the THoR-NP can specifically ablate tumor cells (breast cancer cells) via magnetic hyperthermia, in conjunction with DDC, even in the absence of any exogenous RS supplements. Finally, by incorporating iRGD peptide moieties in the THoR-NP, integrin-enriched cancer cells (malignant tumors, MDA-MB-231) were effectively and selectively killed, as opposed to nonmetastatic tumors (MCF-7), as confirmed in a mouse xenograft model. Hence, our strategy of using nanoparticles embedded with ROS-scavenger-inhibitor with an exogenous ROS supplement is highly selective and effective cancer therapy.
Collapse
Affiliation(s)
- Hyeon-Yeol Cho
- Department of Chemistry and Chemical Biology , Rutgers, The State University of New Jersey , Piscataway , New Jersey 08854 , United States
- Department of Chemical & Biomolecular Engineering , Sogang University , Seoul 04107 , Republic of Korea
| | - Ahmet Mavi
- Department of Nanobiotechnology , Atatürk University , Erzurum 25030 , Turkey
| | - Sy-Tsong Dean Chueng
- Department of Chemistry and Chemical Biology , Rutgers, The State University of New Jersey , Piscataway , New Jersey 08854 , United States
| | - Thanapat Pongkulapa
- Department of Chemistry and Chemical Biology , Rutgers, The State University of New Jersey , Piscataway , New Jersey 08854 , United States
| | - Nicholas Pasquale
- Department of Chemistry and Chemical Biology , Rutgers, The State University of New Jersey , Piscataway , New Jersey 08854 , United States
| | - Hudifah Rabie
- Department of Chemistry and Chemical Biology , Rutgers, The State University of New Jersey , Piscataway , New Jersey 08854 , United States
| | - Jiyou Han
- Department of Biological Sciences, Laboratory of Stem Cell Research and Biotechnology , Hyupsung University , Hwasung-si 18330 , Republic of Korea
| | - Jong Hoon Kim
- Department of Biotechnology, Laboratory of Stem Cells and Tissue Regeneration, College of Life Sciences and Biotechnology , Korea University , Seoul 02841 , Republic of Korea
| | - Tae-Hyung Kim
- School of Integrative Engineering , Chung-Ang University , Seoul 06974 , Republic of Korea
| | - Jeong-Woo Choi
- Department of Chemical & Biomolecular Engineering , Sogang University , Seoul 04107 , Republic of Korea
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology , Rutgers, The State University of New Jersey , Piscataway , New Jersey 08854 , United States
- Department of Life and Nanopharmaceutical Science, College of Pharmacy , Kyung Hee University , Seoul 02447 , Republic of Korea
| |
Collapse
|
5
|
Giniatullin A, Petrov A, Giniatullin R. Action of Hydrogen Peroxide on Synaptic Transmission at the Mouse Neuromuscular Junction. Neuroscience 2018; 399:135-145. [PMID: 30593920 DOI: 10.1016/j.neuroscience.2018.12.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/30/2018] [Accepted: 12/17/2018] [Indexed: 12/15/2022]
Abstract
Hydrogen peroxide (H2O2) is one of the reactive oxygen species (ROS), endogenously produced during metabolism, which acts as a second messenger. In skeletal muscles, hypoxia- or hyperthermia-induced increase in H2O2 might affect synaptic transmission by targeting the most redox-sensitive presynaptic compartment (Giniatullin et al., 2006). However, the effects of H2O2 as a signal molecule have not previously been studied in different patterns of the synaptic activity. Here, using optical and microelectrode recording of synaptic vesicle exocytosis, we studied the use-dependent action of low concentrations of H2O2 and other oxidants in the mouse neuromuscular junction. We found that: (i) H2O2 at low micromole concentrations inhibited both spontaneous and evoked transmitter releases from the motor nerve terminals in a use-dependent manner, (ii) the antioxidant N-acetylcysteine (NAC) eliminated these depressant effects, (iii) the influence of H2O2 was not associated with lipid oxidation suggesting a pure signaling action, (iv) the intracellular oxidant Chloramine-T or (v) the glutathione depletion produced similar to H2O2 depressant effects. Taken together, our data revealed the effective inhibition of neurotransmitter release by ROS, which was proportional to the intensity of synaptic activity at the neuromuscular junction. The combination of various oxidants suggested an intracellular location for redox-sensitive sites responsible for modulation of the synaptic transmission in the skeletal muscle.
Collapse
Affiliation(s)
| | - Alexey Petrov
- Institute of Neuroscience, Kazan State Medial University, Kazan, Russia; Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Centre "Kazan Scientific Centre of RAS", Kazan, Russia
| | - Rashid Giniatullin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia; A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
6
|
Tomkova S, Misuth M, Lenkavska L, Miskovsky P, Huntosova V. In vitro identification of mitochondrial oxidative stress production by time-resolved fluorescence imaging of glioma cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:616-628. [PMID: 29410069 DOI: 10.1016/j.bbamcr.2018.01.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 02/07/2023]
Abstract
Oxidative phosphorylation and glycolysis are important features, by which cells could bypass oxidative stress. The level of oxidative stress, and the ability of cells to promote oxidative phosphorylation or glycolysis, significantly determined proliferation or cell demise. In the present work, we have employed selective mitochondrial probe MitoTracker™ Orange CMTM/Ros (MTO) to estimate the level of oxidative stress in cancer cells at different stressed conditions. MTO is partially sensitive to decrease of mitochondrial membrane potential and to reactive oxygen species (ROS) generated in mitochondria. We have demonstrated, that fluorescence lifetime of MTO is much more sensitive to oxidative stress than intensity-based approaches. This method was validated in different cancer cell lines. Our approach revealed, at relatively low ROS levels, that Gö 6976, a protein kinase C (PKC) α inhibitor, and rottlerin, an indirect PKCδ inhibitor, increased mitochondrial ROS level in glioma cell. Their involvement in oxidative phosphorylation and apoptosis was investigated with oxygen consumption rate estimation, western blot and flow-cytometric analysis. Our study brings new insight to identify feeble differences in ROS production in living cells.
Collapse
Affiliation(s)
- Silvia Tomkova
- Department of Biophysics, Faculty of Science, P. J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia
| | - Matus Misuth
- Department of Biophysics, Faculty of Science, P. J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia
| | - Lenka Lenkavska
- Department of Biophysics, Faculty of Science, P. J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia
| | - Pavol Miskovsky
- Center for Interdisciplinary Biosciences, Technology and innovation park, P.J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia; SAFTRA photonics Ltd., Jesenna 5, 041 54, Kosice, Slovakia
| | - Veronika Huntosova
- Center for Interdisciplinary Biosciences, Technology and innovation park, P.J. Safarik University in Kosice, Jesenna 5, 041 54, Kosice, Slovakia.
| |
Collapse
|
7
|
Novgorodov SA, Voltin JR, Gooz MA, Li L, Lemasters JJ, Gudz TI. Acid sphingomyelinase promotes mitochondrial dysfunction due to glutamate-induced regulated necrosis. J Lipid Res 2017; 59:312-329. [PMID: 29282302 DOI: 10.1194/jlr.m080374] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 12/05/2017] [Indexed: 12/11/2022] Open
Abstract
Inhibiting the glutamate/cystine antiporter system xc-, a key antioxidant defense machinery in the CNS, could trigger a novel form of regulated necrotic cell death, ferroptosis. The underlying mechanisms of system xc--dependent cell demise were elucidated using primary oligodendrocytes (OLs) treated with glutamate to block system xc- function. Pharmacological analysis revealed ferroptosis as a major contributing factor to glutamate-initiated OL death. A sphingolipid profile showed elevations of ceramide species and sphingosine that were preventable by inhibiting of an acid sphingomyelinase (ASM) activity. OL survival was enhanced by both downregulating ASM expression and blocking ASM activity. Glutamate-induced ASM activation seems to involve posttranscriptional mechanisms and was associated with a decreased GSH level. Further investigation of the mechanisms of OL response to glutamate revealed enhanced reactive oxygen species production, augmented lipid peroxidation, and opening of the mitochondrial permeability transition pore that were attenuated by hindering ASM. Of note, knocking down sirtuin 3, a deacetylase governing the mitochondrial antioxidant system, reduced OL survival. The data highlight the importance of the mitochondrial compartment in regulated necrotic cell death and accentuate the novel role of ASM in disturbing mitochondrial functions during OL response to glutamate toxicity, which is essential for pathobiology in stroke and traumatic brain injury.
Collapse
Affiliation(s)
- Sergei A Novgorodov
- Departments of Neuroscience Medical University of South Carolina, Charleston, SC 29425
| | - Joshua R Voltin
- Departments of Neuroscience Medical University of South Carolina, Charleston, SC 29425
| | - Monika A Gooz
- Departments of Drug Discovery, Medical University of South Carolina, Charleston, SC 29425
| | - Li Li
- Departments of Drug Discovery, Medical University of South Carolina, Charleston, SC 29425
| | - John J Lemasters
- Departments of Drug Discovery, Medical University of South Carolina, Charleston, SC 29425
| | - Tatyana I Gudz
- Departments of Neuroscience Medical University of South Carolina, Charleston, SC 29425 .,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401
| |
Collapse
|
8
|
Wang Y, Yao Y, Nie H, He X. Implication of protein kinase C of the left intermediate medial mesopallium in memory impairments induced by early prenatal morphine exposure in one-day old chicks. Eur J Pharmacol 2016; 795:94-100. [PMID: 27940175 DOI: 10.1016/j.ejphar.2016.12.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 12/02/2016] [Accepted: 12/06/2016] [Indexed: 02/06/2023]
Abstract
Previously we reported that prenatal morphine exposure during embryonic days 5-8 can cause cognitive deficits of one-trial passive avoidance learning (PAL) in one-day old chicks. Because protein kinase C (PKC) has been associated with memory capacity, we investigated the effects of prenatal morphine exposure on PKC isoforms expression in the left intermediate medial mesopallium (IMM) of chick brain at a time when memory tests were performed at 30, 120 and 360min respectively following training in PAL paradigm. We found that the level of PKCα in the membrane fractions in left IMM was decreased but that in the cytosol fractions showed a increased trend in prenatally morphine-exposed chicks with impaired long-term memory (120 and 360min). Moreover, the translocation of PKC δ from cytosol to membrane in left IMM was shown in prenatal morphine group which had significantly impaired long-term memory at 360min after training. Furthermore, there were no statistical differences between the two groups regarding the expressions of PKCα and PKC δ in the membrane fraction, although their levels in the cytosol fraction of prenatal morphine group which showed impaired intermediate-term memory at 30min after training, were quite different from that of prenatal saline group. Taken together, these results indicate that PKCα and PKC δ in the left IMM are differentially involved in the impairments of long-term memory induced by prenatal morphine exposure. Neither PKCα nor PKC δ in left IMM may be associated with the disruption of intermediate-term memory of chicks prenatally exposed to morphine.
Collapse
Affiliation(s)
- Ying Wang
- School of Medical Humanities, Tianjin Medical University, Tianjin, PR China
| | - Yang Yao
- Department of Clinical Biochemistry, School of Medical Laboratory, Tianjin Medical University, Tianjin, PR China
| | - Han Nie
- College of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing, PR China
| | - Xingu He
- School of Medical Humanities, Tianjin Medical University, Tianjin, PR China.
| |
Collapse
|
9
|
Mancilla H, Maldonado R, Cereceda K, Villarroel-Espíndola F, Montes de Oca M, Angulo C, Castro MA, Slebe JC, Vera JC, Lavandero S, Concha II. Glutathione Depletion Induces Spermatogonial Cell Autophagy. J Cell Biochem 2016; 116:2283-92. [PMID: 25833220 DOI: 10.1002/jcb.25178] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 03/27/2015] [Indexed: 12/13/2022]
Abstract
The development and survival of male germ cells depend on the antioxidant capacity of the seminiferous tubule. Glutathione (GSH) plays an important role in the antioxidant defenses of the spermatogenic epithelium. Autophagy can act as a pro-survival response during oxidative stress or nutrient deficiency. In this work, we evaluated whether autophagy is involved in spermatogonia-type germ cell survival during severe GSH deficiency. We showed that the disruption of GSH metabolism with l-buthionine-(S,R)-sulfoximine (BSO) decreased reduced (GSH), oxidized (GSSG) glutathione content, and GSH/GSSG ratio in germ cells, without altering reactive oxygen species production and cell viability, evaluated by 2',7'-dichlorodihydrofluorescein (DCF) fluorescence and exclusion of propidium iodide assays, respectively. Autophagy was assessed by processing the endogenous protein LC3I and observing its sub-cellular distribution. Immunoblot and immunofluorescence analysis showed a consistent increase in LC3II and accumulation of autophagic vesicles under GSH-depletion conditions. This condition did not show changes in the level of phosphorylation of AMP-activated protein kinase (AMPK) or the ATP content. A loss in S-glutathionylated protein pattern was also observed. However, inhibition of autophagy resulted in decreased ATP content and increased caspase-3/7 activity in GSH-depleted germ cells. These findings suggest that GSH deficiency triggers an AMPK-independent induction of autophagy in germ cells as an adaptive stress response.
Collapse
Affiliation(s)
- Héctor Mancilla
- Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia, Chile
| | - Rodrigo Maldonado
- Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia, Chile
| | - Karina Cereceda
- Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia, Chile
| | | | - Marco Montes de Oca
- Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia, Chile
| | - Constanza Angulo
- Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia, Chile
| | - Maite A Castro
- Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia, Chile
| | - Juan C Slebe
- Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia, Chile
| | - Juan C Vera
- Departamento de Fisiopatología, Universidad de Concepción, Concepción, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS) & Center for Molecular Studies of the Cell, Facultad Ciencias Químicas y Farmacéuticas & Facultad Medicina, Universidad de Chile, Santiago, Chile
| | - Ilona I Concha
- Instituto de Bioquímica y Microbiología, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
10
|
Kornfeld OS, Hwang S, Disatnik MH, Chen CH, Qvit N, Mochly-Rosen D. Mitochondrial reactive oxygen species at the heart of the matter: new therapeutic approaches for cardiovascular diseases. Circ Res 2015; 116:1783-99. [PMID: 25999419 DOI: 10.1161/circresaha.116.305432] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Reactive oxygen species (ROS) have been implicated in a variety of age-related diseases, including multiple cardiovascular disorders. However, translation of ROS scavengers (antioxidants) into the clinic has not been successful. These antioxidants grossly reduce total levels of cellular ROS including ROS that participate in physiological signaling. In this review, we challenge the traditional antioxidant therapeutic approach that targets ROS directly with novel approaches that improve mitochondrial functions to more effectively treat cardiovascular diseases.
Collapse
Affiliation(s)
- Opher S Kornfeld
- From the Department of Chemical and Systems Biology, Stanford University School of Medicine, CA
| | - Sunhee Hwang
- From the Department of Chemical and Systems Biology, Stanford University School of Medicine, CA
| | - Marie-Hélène Disatnik
- From the Department of Chemical and Systems Biology, Stanford University School of Medicine, CA
| | - Che-Hong Chen
- From the Department of Chemical and Systems Biology, Stanford University School of Medicine, CA
| | - Nir Qvit
- From the Department of Chemical and Systems Biology, Stanford University School of Medicine, CA
| | - Daria Mochly-Rosen
- From the Department of Chemical and Systems Biology, Stanford University School of Medicine, CA.
| |
Collapse
|
11
|
Lucke-Wold BP, Turner RC, Logsdon AF, Simpkins JW, Alkon DL, Smith KE, Chen YW, Tan Z, Huber JD, Rosen CL. Common mechanisms of Alzheimer's disease and ischemic stroke: the role of protein kinase C in the progression of age-related neurodegeneration. J Alzheimers Dis 2015; 43:711-24. [PMID: 25114088 PMCID: PMC4446718 DOI: 10.3233/jad-141422] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Ischemic stroke and Alzheimer's disease (AD), despite being distinct disease entities, share numerous pathophysiological mechanisms such as those mediated by inflammation, immune exhaustion, and neurovascular unit compromise. An important shared mechanistic link is acute and chronic changes in protein kinase C (PKC) activity. PKC isoforms have widespread functions important for memory, blood-brain barrier maintenance, and injury repair that change as the body ages. Disease states accelerate PKC functional modifications. Mutated forms of PKC can contribute to neurodegeneration and cognitive decline. In some cases the PKC isoforms are still functional but are not successfully translocated to appropriate locations within the cell. The deficits in proper PKC translocation worsen stroke outcome and amyloid-β toxicity. Cross talk between the innate immune system and PKC pathways contribute to the vascular status within the aging brain. Unfortunately, comorbidities such as diabetes, obesity, and hypertension disrupt normal communication between the two systems. The focus of this review is to highlight what is known about PKC function, how isoforms of PKC change with age, and what additional alterations are consequences of stroke and AD. The goal is to highlight future therapeutic targets that can be applied to both the treatment and prevention of neurologic disease. Although the pathology of ischemic stroke and AD are different, the similarity in PKC responses warrants further investigation, especially as PKC-dependent events may serve as an important connection linking age-related brain injury.
Collapse
Affiliation(s)
- Brandon P. Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Ryan C. Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Aric F. Logsdon
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, USA
| | - James W. Simpkins
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Daniel L. Alkon
- Blanchette Rockefeller Neurosciences Institute, Morgantown, WV, USA
| | - Kelly E. Smith
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, USA
| | - Yi-Wen Chen
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Zhenjun Tan
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Jason D. Huber
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, USA
| | - Charles L. Rosen
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- Correspondence to: Charles L. Rosen, MD, PhD, Department of Neurosurgery, West Virginia University School of Medicine, One Medical Center Drive, Suite 4300, Health Sciences Center, PO Box 9183, Morgantown, WV 26506-9183, USA. Tel.: +1 304 293 5041; Fax: +1 304 293 4819;
| |
Collapse
|
12
|
Jeong KW. Flightless I (Drosophila) homolog facilitates chromatin accessibility of the estrogen receptor α target genes in MCF-7 breast cancer cells. Biochem Biophys Res Commun 2014; 446:608-13. [PMID: 24632205 DOI: 10.1016/j.bbrc.2014.03.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 03/04/2014] [Indexed: 12/13/2022]
Abstract
The coordinated activities of multiple protein complexes are essential to the remodeling of chromatin structure and for the recruitment of RNA polymerase II (Pol II) to the promoter in order to facilitate the initiation of transcription in nuclear receptor-mediated gene expression. Flightless I (Drosophila) homolog (FLII), a nuclear receptor coactivator, is associated with the SWI/SNF-chromatin remodeling complex during estrogen receptor (ER)α-mediated transcription. However, the function of FLII in estrogen-induced chromatin opening has not been fully explored. Here, we show that FLII plays a critical role in establishing active histone modification marks and generating the open chromatin structure of ERα target genes. We observed that the enhancer regions of ERα target genes are heavily occupied by FLII, and histone H3K4me3 and Pol II binding induced by estrogen are decreased in FLII-depleted MCF-7 cells. Furthermore, formaldehyde-assisted isolation of regulatory elements (FAIRE)-quantitative polymerase chain reaction (qPCR) experiments showed that depletion of FLII resulted in reduced chromatin accessibility of multiple ERα target genes. These data suggest FLII as a key regulator of ERα-mediated transcription through its role in regulating chromatin accessibility for the binding of RNA Polymerase II and possibly other transcriptional coactivators.
Collapse
Affiliation(s)
- Kwang Won Jeong
- Gachon Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, 7-45 Songdo-dong, Yeonsu-gu, Incheon 406-840, Republic of Korea.
| |
Collapse
|