151
|
Ballinger MA, Schwartz C, Andrews MT. Enhanced oxidative capacity of ground squirrel brain mitochondria during hibernation. Am J Physiol Regul Integr Comp Physiol 2017; 312:R301-R310. [PMID: 28077389 DOI: 10.1152/ajpregu.00314.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 01/06/2017] [Accepted: 01/08/2017] [Indexed: 11/22/2022]
Abstract
During hibernation, thirteen-lined ground squirrels (Ictidomys tridecemlineatus) regularly cycle between bouts of torpor and interbout arousal (IBA). Most of the brain is electrically quiescent during torpor but regains activity quickly upon arousal to IBA, resulting in extreme oscillations in energy demand during hibernation. We predicted increased functional capacity of brain mitochondria during hibernation compared with spring to accommodate the variable energy demands of hibernation. To address this hypothesis, we examined mitochondrial bioenergetics in the ground squirrel brain across three time points: spring (SP), torpor (TOR), and IBA. Respiration rates of isolated brain mitochondria through complex I of the electron transport chain were more than twofold higher in TOR and IBA than in SP (P < 0.05). We also found a 10% increase in membrane potential between hibernation and spring (P < 0.05), and that proton leak was lower in TOR and IBA than in SP. Finally, there was a 30% increase in calcium loading in SP brain mitochondria compared with TOR and IBA (P < 0.01). To analyze brain mitochondrial abundance between spring and hibernation, we measured the ratio of copy number in a mitochondrial gene (ND1) vs. a nuclear gene (B2M) in frozen cerebral cortex samples. No significant differences were observed in DNA copies between SP and IBA. These data show that brain mitochondrial bioenergetics are not static across the year and suggest that brain mitochondria function more effectively during the hibernation season, allowing for rapid production of energy to meet demand when extreme physiological changes are occurring.
Collapse
Affiliation(s)
- Mallory A Ballinger
- Department of Biology, University of Minnesota Duluth, Duluth, Minnesota; and
| | - Christine Schwartz
- Department of Biology, University of Minnesota Duluth, Duluth, Minnesota; and.,Department of Biology, University of Wisconsin-La Crosse, La Crosse, Wisconsin
| | - Matthew T Andrews
- Department of Biology, University of Minnesota Duluth, Duluth, Minnesota; and
| |
Collapse
|
152
|
Lomeli N, Di K, Czerniawski J, Guzowski JF, Bota DA. Cisplatin-induced mitochondrial dysfunction is associated with impaired cognitive function in rats. Free Radic Biol Med 2017; 102:274-286. [PMID: 27908784 PMCID: PMC5308450 DOI: 10.1016/j.freeradbiomed.2016.11.046] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/22/2016] [Accepted: 11/27/2016] [Indexed: 02/08/2023]
Abstract
PURPOSE Chemotherapy-related cognitive impairment (CRCI) is commonly reported following the administration of chemotherapeutic agents and comprises a wide variety of neurological problems. No effective treatments for CRCI are currently available. Here we examined the mechanisms involving cisplatin-induced hippocampal damage following cisplatin administration in a rat model and in cultured rat hippocampal neurons and neural stem/progenitor cells (NSCs). We also assessed the protective effects of the antioxidant, N-acetylcysteine in mitigating these damages. EXPERIMENTAL DESIGN Adult male rats received 6mg/kg cisplatin in the acute studies. In chronic studies, rats received 5mg/kg cisplatin or saline injections once per week for 4 weeks. N-acetylcysteine (250mg/kg/day) or saline was administered for five consecutive days during cisplatin treatment. Cognitive testing was performed 5 weeks after treatment cessation. Cisplatin-treated cultured hippocampal neurons and NSCs were examined for changes in mitochondrial function, oxidative stress production, caspase-9 activation, and neuronal dendritic spine density. RESULTS Acute cisplatin treatment reduced dendritic branching and spine density, and induced mitochondrial degradation. Rats receiving the chronic cisplatin regimen showed impaired performance in contextual fear conditioning, context object discrimination, and novel object recognition tasks compared to controls. Cisplatin induced mitochondrial DNA damage, impaired respiratory activity, increased oxidative stress, and activated caspase-9 in cultured hippocampal neurons and NSCs. N-acetylcysteine treatment prevented free radical production, ameliorated apoptotic cellular death and dendritic spine loss, and partially reversed the cisplatin-induced cognitive impairments. CONCLUSIONS Our results suggest that mitochondrial dysfunction and increased oxidative stress are involved in cisplatin-induced cognitive impairments. Therapeutic agents, such as N-acetylcysteine, may be effective in mitigating the deleterious effects of cisplatin.
Collapse
Affiliation(s)
- Naomi Lomeli
- Department of Pathology & Laboratory Medicine, University of California Irvine, Irvine, CA, USA.
| | - Kaijun Di
- Department of Neurological Surgery, University of California Irvine, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA.
| | - Jennifer Czerniawski
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA, USA; Center for the Neurobiology of Learning & Memory, University of California Irvine, Irvine, CA, USA.
| | - John F Guzowski
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA, USA; Center for the Neurobiology of Learning & Memory, University of California Irvine, Irvine, CA, USA.
| | - Daniela A Bota
- Department of Pathology & Laboratory Medicine, University of California Irvine, Irvine, CA, USA; Department of Neurological Surgery, University of California Irvine, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA; Department of Neurology, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
153
|
Yao C, Song H, Wan Y, Ma K, Zheng C, Cui H, Xin P, Ji X, Deng S. Electro-Photodynamic Visualization of Singlet Oxygen Induced by Zinc Porphyrin Modified Microchip in Aqueous Media. ACS APPLIED MATERIALS & INTERFACES 2016; 8:34833-34843. [PMID: 27762540 DOI: 10.1021/acsami.6b10213] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
A porphyrin-based electro-photodynamic imaging system was fabricated for monitoring the concentration of oxygen. Distinct from the electrochemiluminescent (ECL) inability of numerous organic species in aqueous solutions, a strong and stable red irradiation at 634 nm could be stimulated electrochemically on zinc(II) meso-tetra(4-carboxyphenyl) porphine (ZnTCPP)/tetraoctylammonium bromide (TOAB) in the physiological condition. In terms of in situ electron paramagnetic resonance and ECL spectroscopies, the nature of ECL was thoroughly investigated, being exactly the chemiluminescence from singlet oxygen (1O2) produced during the successive electro-reduction of ZnTCPP. Meanwhile, the excellent film-making capacity of amphiphilic TOAB as a potent ion barrier granted the luminophores a micro-order and patternable electrode modification. Such platform was exceptionally tolerant of pH variation, facilitating a durable solid-state ECL visualization under potentiostatic electrolysis and time exposure in the charge-coupled device (CCD) camera. For flow-injection and real-time detection, a chip-mounted microfluidic cell was customized and manufactured. A sensitive and simple vision-sensing of O2 was further achieved with a real determination limit as low as a few micromolar level. The developed ECL imaging system is a good prototype and an eco-friendly technique in the cathodic range, and thus, it would supplement the primary anodic imaging library, showing great promise for multiplexed and colorimetric assays as well as oxygen-involved activity studies in the future.
Collapse
Affiliation(s)
- Chuanguang Yao
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology , Nanjing 210094, P.R. China
| | - Hongxin Song
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology , Nanjing 210094, P.R. China
| | - Ying Wan
- Intelligent Microsystem Technology and Engineering Center, School of Mechanical Engineering, Nanjing University of Science and Technology , Nanjing 210094, P.R. China
| | - Kefeng Ma
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology , Nanjing 210094, P.R. China
| | - Chenyu Zheng
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology , Nanjing 210094, P.R. China
| | - Hongda Cui
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology , Nanjing 210094, P.R. China
| | - Peng Xin
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology , Nanjing 210094, P.R. China
| | - Xubo Ji
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology , Nanjing 210094, P.R. China
| | - Shengyuan Deng
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology , Nanjing 210094, P.R. China
- Materials Research Science and Engineering Centers (MRSEC), Department of Chemistry, Penn State University , University Park, Pennsylvania 16801, United States
| |
Collapse
|
154
|
Li R, Steyn FJ, Stout MB, Lee K, Cully TR, Calderón JC, Ngo ST. Development of a high-throughput method for real-time assessment of cellular metabolism in intact long skeletal muscle fibre bundles. J Physiol 2016; 594:7197-7213. [PMID: 27619319 DOI: 10.1113/jp272988] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 09/07/2016] [Indexed: 12/28/2022] Open
Abstract
KEY POINTS We developed a method that allows for real-time assessment of cellular metabolism in isolated, intact long skeletal muscle fibre bundles from adult mice. This method can be used to study changes in mitochondrial function and fuel utilisation in live skeletal muscle fibre bundles. Our method enables flexibility in experimental design and high-throughput assessment of mitochondrial parameters in isolated skeletal muscle fibre bundles. Extensor digitorum longus (EDL) fibre bundles obtained from chronic high-fat diet fed mice had lower basal oxygen consumption under FCCP-induced maximal respiration, when compared to control chow-fed mice. EDL fibre bundles obtained from chronic high-fat diet fed mice had enhanced mitochondrial oxidation capacity under FCCP-induced maximal respiration, when compared to control chow-fed mice. ABSTRACT Metabolic dysfunction in skeletal muscle contributes to the aetiology and development of muscle diseases and metabolic diseases. As such, assessment of skeletal muscle cellular bioenergetics provides a powerful means to understand the role of skeletal muscle metabolism in disease and to identify possible therapeutic targets. Here, we developed a method that allows for the real-time assessment of cellular respiration in intact skeletal muscle fibre bundles obtained from the extensor digitorum longus (EDL) muscle of adult mice. Using this method, we assessed the contribution of ATP turnover and proton leak to basal mitochondrial oxygen consumption rate (OCR). Our data demonstrate that the mitochondria in EDL fibres are loosely coupled. Moreover, in the presence of carbonyl cyanide-p-trifluoromethoxyphenylhydrazone (FCCP), we show that palmitate exposure induced comparable peak OCR and higher total OCR in EDL fibre bundles when compared to pyruvate exposure, suggesting that fatty acids might be a more sustainable fuel source for skeletal muscle when mitochondria are driven to maximal respiration. Application of this method to EDL fibre bundles obtained from chronic high-fat diet fed mice revealed lower basal OCR and enhanced mitochondrial oxidation capacity in the presence of FCCP when compared to the chow-diet fed control mice. By using a 96-well microplate format, our method provides a flexible and efficient platform to investigate mitochondrial parameters of intact skeletal muscle fibres obtained from adult mice.
Collapse
Affiliation(s)
- Rui Li
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Frederik J Steyn
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia.,University of Queensland Centre for Clinical Research, Brisbane, Australia
| | - Michael B Stout
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Kevin Lee
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Tanya R Cully
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Juan C Calderón
- Department of Physiology and Biochemistry, Physiology and Biochemistry Research Group-PHYSIS, Faculty of Medicine, University of Antioquia, Medellín, Colombia
| | - Shyuan T Ngo
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia.,University of Queensland Centre for Clinical Research, Brisbane, Australia.,Queensland Brain Institute, University of Queensland, Brisbane, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Australia
| |
Collapse
|
155
|
Levy G, Habib N, Guzzardi MA, Kitsberg D, Bomze D, Ezra E, Uygun BE, Uygun K, Trippler M, Schlaak JF, Shibolet O, Sklan EH, Cohen M, Timm J, Friedman N, Nahmias Y. Nuclear receptors control pro-viral and antiviral metabolic responses to hepatitis C virus infection. Nat Chem Biol 2016; 12:1037-1045. [PMID: 27723751 DOI: 10.1038/nchembio.2193] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 08/02/2016] [Indexed: 12/11/2022]
Abstract
Viruses lack the basic machinery needed to replicate and therefore must hijack the host's metabolism to propagate. Virus-induced metabolic changes have yet to be systematically studied in the context of host transcriptional regulation, and such studies shoul offer insight into host-pathogen metabolic interplay. In this work we identified hepatitis C virus (HCV)-responsive regulators by coupling system-wide metabolic-flux analysis with targeted perturbation of nuclear receptors in primary human hepatocytes. We found HCV-induced upregulation of glycolysis, ketogenesis and drug metabolism, with glycolysis controlled by activation of HNF4α, ketogenesis by PPARα and FXR, and drug metabolism by PXR. Pharmaceutical inhibition of HNF4α reversed HCV-induced glycolysis, blocking viral replication while increasing apoptosis in infected cells showing virus-induced dependence on glycolysis. In contrast, pharmaceutical inhibition of PPARα or FXR reversed HCV-induced ketogenesis but increased viral replication, demonstrating a novel host antiviral response. Our results show that virus-induced changes to a host's metabolism can be detrimental to its life cycle, thus revealing a biologically complex relationship between virus and host.
Collapse
Affiliation(s)
- Gahl Levy
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.,Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Naomi Habib
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.,Grass Center for Bioengineering, Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.,Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Maria Angela Guzzardi
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.,Institute of Clinical Physiology, National Research Council (CNR), Pisa, Italy
| | - Daniel Kitsberg
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.,Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - David Bomze
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.,Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Elishai Ezra
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.,Faculty of Engineering, Jerusalem College of Technology, Jerusalem, Israel
| | - Basak E Uygun
- Center for Engineering in Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Korkut Uygun
- Center for Engineering in Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Martin Trippler
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Joerg F Schlaak
- Department of Gastroenterology and Hepatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Oren Shibolet
- Liver Unit, Department of Gastroenterology, Tel-Aviv Medical Center and Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Ella H Sklan
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Merav Cohen
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.,Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Joerg Timm
- Institute for Virology, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - Nir Friedman
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.,Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yaakov Nahmias
- Grass Center for Bioengineering, Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.,Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
156
|
Lizama-Manibusan BN, Klein S, McKenzie JR, Cliffel DE, McLaughlin B. Analysis of a Nitroreductase-Based Hypoxia Sensor in Primary Neuronal Cultures. ACS Chem Neurosci 2016; 7:1188-91. [PMID: 27504748 PMCID: PMC5125302 DOI: 10.1021/acschemneuro.6b00215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The ability to assess oxygenation within living cells is much sought after to more deeply understand normal and pathological cell biology. Hypoxia Red manufactured by Enzo Life Sciences is advertised as a novel hypoxia detector dependent on nitroreducatase activity. We sought to use Hypoxia Red in primary neuronal cultures to test cell-to-cell metabolic variability in response to hypoxic stress. Neurons treated with 90 min of hypoxia were labeled with Hypoxia Red. We observed that, even under normoxic conditions neurons expressed fluorescence robustly. Analysis of the chemical reactions and biological underpinnings of this method revealed that the high uptake and reduction of the dye is due to active nitroreductases in normoxic cells that are independent of oxygen availability.
Collapse
Affiliation(s)
| | - Sharon Klein
- Neurology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Jennifer R. McKenzie
- Departments of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - David E. Cliffel
- Departments of Chemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - BethAnn McLaughlin
- Neurology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
| |
Collapse
|
157
|
TeSlaa T, Chaikovsky AC, Lipchina I, Escobar SL, Hochedlinger K, Huang J, Graeber TG, Braas D, Teitell MA. α-Ketoglutarate Accelerates the Initial Differentiation of Primed Human Pluripotent Stem Cells. Cell Metab 2016; 24:485-493. [PMID: 27476976 PMCID: PMC5023506 DOI: 10.1016/j.cmet.2016.07.002] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 02/20/2016] [Accepted: 07/01/2016] [Indexed: 02/06/2023]
Abstract
Pluripotent stem cells (PSCs) can self-renew or differentiate from naive or more differentiated, primed, pluripotent states established by specific culture conditions. Increased intracellular α-ketoglutarate (αKG) was shown to favor self-renewal in naive mouse embryonic stem cells (mESCs). The effect of αKG or αKG/succinate levels on differentiation from primed human PSCs (hPSCs) or mouse epiblast stem cells (EpiSCs) remains unknown. We examined primed hPSCs and EpiSCs and show that increased αKG or αKG-to-succinate ratios accelerate, and elevated succinate levels delay, primed PSC differentiation. αKG has been shown to inhibit the mitochondrial ATP synthase and to regulate epigenome-modifying dioxygenase enzymes. Mitochondrial uncoupling did not impede αKG-accelerated primed PSC differentiation. Instead, αKG induced, and succinate impaired, global histone and DNA demethylation in primed PSCs. The data support αKG promotion of self-renewal or differentiation depending on the pluripotent state.
Collapse
Affiliation(s)
- Tara TeSlaa
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Andrea C Chaikovsky
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Inna Lipchina
- Department of Molecular Biology, Cancer Center and Center for Regenerative Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Sandra L Escobar
- Department of Biology, California State University, Northridge, Northridge, CA 91330, USA
| | - Konrad Hochedlinger
- Department of Molecular Biology, Cancer Center and Center for Regenerative Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Jing Huang
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Thomas G Graeber
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA Metabolomics Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Daniel Braas
- Department of Molecular and Medical Pharmacology, Crump Institute for Molecular Imaging, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA Metabolomics Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michael A Teitell
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, Department of Pediatrics, and Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
158
|
Lamprecht DA, Finin PM, Rahman MA, Cumming BM, Russell SL, Jonnala SR, Adamson JH, Steyn AJC. Turning the respiratory flexibility of Mycobacterium tuberculosis against itself. Nat Commun 2016; 7:12393. [PMID: 27506290 PMCID: PMC4987515 DOI: 10.1038/ncomms12393] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 06/28/2016] [Indexed: 11/21/2022] Open
Abstract
The Mycobacterium tuberculosis (Mtb) electron transport chain (ETC) has received significant attention as a drug target, however its vulnerability may be affected by its flexibility in response to disruption. Here we determine the effect of the ETC inhibitors bedaquiline, Q203 and clofazimine on the Mtb ETC, and the value of the ETC as a drug target, by measuring Mtb's respiration using extracellular flux technology. We find that Mtb's ETC rapidly reroutes around inhibition by these drugs and increases total respiration to maintain ATP levels. Rerouting is possible because Mtb rapidly switches between terminal oxidases, and, unlike eukaryotes, is not susceptible to back pressure. Increased ETC activity potentiates clofazimine's production of reactive oxygen species, causing rapid killing in vitro and in a macrophage model. Our results indicate that combination therapy targeting the ETC can be exploited to enhance killing of Mtb.
Collapse
Affiliation(s)
- Dirk A. Lamprecht
- KwaZulu Natal Research Institute for Tuberculosis and HIV (K-RITH), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban 4001, South Africa
| | - Peter M. Finin
- KwaZulu Natal Research Institute for Tuberculosis and HIV (K-RITH), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban 4001, South Africa
- Department of Internal Medicine, University of Pittsburgh, 1218 Scaife Hall 3550 Terrace Street, Pittsburgh, Pennsylvania 15261, USA
| | - Md. Aejazur Rahman
- KwaZulu Natal Research Institute for Tuberculosis and HIV (K-RITH), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban 4001, South Africa
| | - Bridgette M. Cumming
- KwaZulu Natal Research Institute for Tuberculosis and HIV (K-RITH), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban 4001, South Africa
| | - Shannon L. Russell
- KwaZulu Natal Research Institute for Tuberculosis and HIV (K-RITH), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban 4001, South Africa
| | | | - John H. Adamson
- KwaZulu Natal Research Institute for Tuberculosis and HIV (K-RITH), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban 4001, South Africa
| | - Adrie J. C. Steyn
- KwaZulu Natal Research Institute for Tuberculosis and HIV (K-RITH), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban 4001, South Africa
- Department of Microbiology, University of Alabama at Birmingham, 1720 2nd Avenue South, Birmingham, Alabama 35294-2170, USA
- Centres for AIDS Research and Free Radical Biology, University of Alabama at Birmingham, 1720 2nd Avenue South, Birmingham, Alabama 35294-2170, USA
| |
Collapse
|
159
|
Super A, Jaccard N, Cardoso Marques MP, Macown RJ, Griffin LD, Veraitch FS, Szita N. Real-time monitoring of specific oxygen uptake rates of embryonic stem cells in a microfluidic cell culture device. Biotechnol J 2016; 11:1179-89. [PMID: 27214658 PMCID: PMC5103178 DOI: 10.1002/biot.201500479] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 02/15/2016] [Accepted: 05/12/2016] [Indexed: 01/07/2023]
Abstract
Oxygen plays a key role in stem cell biology as a signaling molecule and as an indicator of cell energy metabolism. Quantification of cellular oxygen kinetics, i.e. the determination of specific oxygen uptake rates (sOURs), is routinely used to understand metabolic shifts. However current methods to determine sOUR in adherent cell cultures rely on cell sampling, which impacts on cellular phenotype. We present real‐time monitoring of cell growth from phase contrast microscopy images, and of respiration using optical sensors for dissolved oxygen. Time‐course data for bulk and peri‐cellular oxygen concentrations obtained for Chinese hamster ovary (CHO) and mouse embryonic stem cell (mESCs) cultures successfully demonstrated this non‐invasive and label‐free approach. Additionally, we confirmed non‐invasive detection of cellular responses to rapidly changing culture conditions by exposing the cells to mitochondrial inhibiting and uncoupling agents. For the CHO and mESCs, sOUR values between 8 and 60 amol cell−1 s−1, and 5 and 35 amol cell−1 s−1 were obtained, respectively. These values compare favorably with literature data. The capability to monitor oxygen tensions, cell growth, and sOUR, of adherent stem cell cultures, non‐invasively and in real time, will be of significant benefit for future studies in stem cell biology and stem cell‐based therapies.
Collapse
Affiliation(s)
- Alexandre Super
- Department of Biochemical Engineering, University College London, London, United Kingdom
| | - Nicolas Jaccard
- Department of Biochemical Engineering, University College London, London, United Kingdom.,Centre for Mathematics and Physics in the Life Sciences and Experimental Biology, University College London, London, United Kingdom.,Department of Computer Science, University College London, London, United Kingdom
| | | | - Rhys Jarred Macown
- Department of Biochemical Engineering, University College London, London, United Kingdom
| | - Lewis Donald Griffin
- Department of Computer Science, University College London, London, United Kingdom
| | - Farlan Singh Veraitch
- Department of Biochemical Engineering, University College London, London, United Kingdom
| | - Nicolas Szita
- Department of Biochemical Engineering, University College London, London, United Kingdom.
| |
Collapse
|
160
|
Aw WC, Bajracharya R, Towarnicki SG, Ballard JWO. Assessing bioenergetic functions from isolated mitochondria in Drosophila melanogaster. J Biol Methods 2016; 3:e42. [PMID: 31453209 PMCID: PMC6706135 DOI: 10.14440/jbm.2016.112] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/17/2016] [Accepted: 05/16/2016] [Indexed: 11/23/2022] Open
Abstract
Mitochondria are involved in generating more than 90 percent of cellular energy and are responsible for many cellular processes such as metabolism, cell signalling, apoptosis and ageing. Currently, there are a number of different experimental approaches employed to measure mitochondrial health and function. Here, we demonstrate a novel approach that quantifies substrate induced mitochondrial respiration from Drosophila. This protocol is optimized for mitochondria isolated from third instar larvae, and can also be used for mitochondria isolated from adult thoraces. This procedure outlines how to perform high throughput and high resolution mitochondria specific measurements for state II, state III, state IVO respiration and residual oxygen consumption.
Collapse
Affiliation(s)
- Wen C Aw
- School of Biotechnology and Biomolecular Science, University of New South Wales, Sydney 2052, Australia
| | - Rijan Bajracharya
- School of Biotechnology and Biomolecular Science, University of New South Wales, Sydney 2052, Australia
| | - Samuel G Towarnicki
- School of Biotechnology and Biomolecular Science, University of New South Wales, Sydney 2052, Australia
| | - J William O Ballard
- School of Biotechnology and Biomolecular Science, University of New South Wales, Sydney 2052, Australia
| |
Collapse
|
161
|
Nwosu ZC, Alborzinia H, Wölfl S, Dooley S, Liu Y. Evolving Insights on Metabolism, Autophagy, and Epigenetics in Liver Myofibroblasts. Front Physiol 2016; 7:191. [PMID: 27313533 PMCID: PMC4887492 DOI: 10.3389/fphys.2016.00191] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 05/12/2016] [Indexed: 12/14/2022] Open
Abstract
Liver myofibroblasts (MFB) are crucial mediators of extracellular matrix (ECM) deposition in liver fibrosis. They arise mainly from hepatic stellate cells (HSCs) upon a process termed “activation.” To a lesser extent, and depending on the cause of liver damage, portal fibroblasts, mesothelial cells, and fibrocytes may also contribute to the MFB population. Targeting MFB to reduce liver fibrosis is currently an area of intense research. Unfortunately, a clog in the wheel of antifibrotic therapies is the fact that although MFB are known to mediate scar formation, and participate in liver inflammatory response, many of their molecular portraits are currently unknown. In this review, we discuss recent understanding of MFB in health and diseases, focusing specifically on three evolving research fields: metabolism, autophagy, and epigenetics. We have emphasized on therapeutic prospects where applicable and mentioned techniques for use in MFB studies. Subsequently, we highlighted uncharted territories in MFB research to help direct future efforts aimed at bridging gaps in current knowledge.
Collapse
Affiliation(s)
- Zeribe C Nwosu
- Molecular Hepatology Section, Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg Mannheim, Germany
| | - Hamed Alborzinia
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg Heidelberg, Germany
| | - Stefan Wölfl
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg Heidelberg, Germany
| | - Steven Dooley
- Molecular Hepatology Section, Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg Mannheim, Germany
| | - Yan Liu
- Molecular Hepatology Section, Department of Medicine II, Medical Faculty Mannheim, University of Heidelberg Mannheim, Germany
| |
Collapse
|
162
|
Ballinger MA, Hess C, Napolitano MW, Bjork JA, Andrews MT. Seasonal changes in brown adipose tissue mitochondria in a mammalian hibernator: from gene expression to function. Am J Physiol Regul Integr Comp Physiol 2016; 311:R325-36. [PMID: 27225952 DOI: 10.1152/ajpregu.00463.2015] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 05/24/2016] [Indexed: 12/25/2022]
Abstract
Brown adipose tissue (BAT) is a thermogenic organ that is vital for hibernation in mammals. Throughout the hibernation season, BAT mitochondrial uncoupling protein 1 (UCP1) enables rapid rewarming from hypothermic torpor to periodic interbout arousals (IBAs), as energy is dissipated as heat. However, BAT's unique ability to rewarm the body via nonshivering thermogenesis is not necessary outside the hibernation season, suggesting a potential seasonal change in the regulation of BAT function. Here, we examined the BAT mitochondrial proteome and mitochondrial bioenergetics in the thirteen-lined ground squirrel (Ictidomys tridecemlineatus) across four time points: spring, fall, torpor, and IBA. Relative mitochondrial content of BAT was estimated by measuring BAT pad mass, UCP1 protein content, and mitochondrial DNA (mtDNA) copy number. BAT mtDNA content was significantly lower in spring compared with torpor and IBA (P < 0.05). UCP1 mRNA and protein levels were highest during torpor and IBA. Respiration rates of isolated BAT mitochondria were interrogated at each complex of the electron transport chain. Respiration at complex II was significantly higher in torpor and IBA compared with spring (P < 0.05), suggesting an enhancement in mitochondrial respiratory capacity during hibernation. Additionally, proteomic iTRAQ labeling identified 778 BAT mitochondrial proteins. Proteins required for mitochondrial lipid translocation and β-oxidation were upregulated during torpor and IBA and downregulated in spring. These data imply that BAT bioenergetics and mitochondrial content are not static across the year, despite the year-round presence of UCP1.
Collapse
Affiliation(s)
| | - Clair Hess
- Department of Biology, University of Minnesota Duluth, Duluth, Minnesota
| | - Max W Napolitano
- Department of Biology, University of Minnesota Duluth, Duluth, Minnesota
| | - James A Bjork
- Department of Biology, University of Minnesota Duluth, Duluth, Minnesota
| | - Matthew T Andrews
- Department of Biology, University of Minnesota Duluth, Duluth, Minnesota
| |
Collapse
|
163
|
Gunda V, Yu F, Singh PK. Validation of Metabolic Alterations in Microscale Cell Culture Lysates Using Hydrophilic Interaction Liquid Chromatography (HILIC)-Tandem Mass Spectrometry-Based Metabolomics. PLoS One 2016; 11:e0154416. [PMID: 27120458 PMCID: PMC4847783 DOI: 10.1371/journal.pone.0154416] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 04/12/2016] [Indexed: 02/04/2023] Open
Abstract
By standard convention, in order to increase the efficacy of metabolite detection from cell culture lysates, metabolite extracts from a large quantity of cells are utilized for multiple reaction monitoring-based metabolomic studies. Metabolomics from a small number of cell extracts offers a potential economical alternative to increased cell numbers, in turn increasing the utility of cell culture-based metabolomics. However, the effect of reduced cell numbers on targeted metabolomic profiling is relatively unstudied. Considering the limited knowledge available of the feasibility and accuracy of microscale cell culture metabolomics, the present study analyzes differences in metabolomic profiles of different cell numbers of three pancreatic cancer cell lines. Specifically, it examines the effects of reduced cell numbers on metabolite profiles by obtaining extracts either directly from microscale culture plates or through serial dilution of increased numbers of cellular metabolite extracts. Our results indicate reduced cell numbers only modestly affect the number of metabolites detected (93% of metabolites detected in cell numbers as low as 104 cells and 97% for 105 cells), independent of the method used to obtain the cells. However, metabolite peak intensities were differentially affected by the reduced cell numbers, with some peak intensities inversely proportional to the cell numbers. To help eliminate such potential inverse relationships, peak intensities for increased cell numbers were excluded from the comparative analysis. Overall, metabolite profiles from microscale culture plates were observed to differ from the serial dilution samples, which may be attributable to the medium-to-cell-number ratios. Finally, findings identify perturbations in metabolomic profiling for cellular extracts from reduced cell numbers, which offer future applications in microscale metabolomic evaluations.
Collapse
Affiliation(s)
- Venugopal Gunda
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Fang Yu
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Pankaj K. Singh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
164
|
Real-time monitoring of metabolic function in liver-on-chip microdevices tracks the dynamics of mitochondrial dysfunction. Proc Natl Acad Sci U S A 2016; 113:E2231-40. [PMID: 27044092 DOI: 10.1073/pnas.1522556113] [Citation(s) in RCA: 212] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Microfluidic organ-on-a-chip technology aims to replace animal toxicity testing, but thus far has demonstrated few advantages over traditional methods. Mitochondrial dysfunction plays a critical role in the development of chemical and pharmaceutical toxicity, as well as pluripotency and disease processes. However, current methods to evaluate mitochondrial activity still rely on end-point assays, resulting in limited kinetic and prognostic information. Here, we present a liver-on-chip device capable of maintaining human tissue for over a month in vitro under physiological conditions. Mitochondrial respiration was monitored in real time using two-frequency phase modulation of tissue-embedded phosphorescent microprobes. A computer-controlled microfluidic switchboard allowed contiguous electrochemical measurements of glucose and lactate, providing real-time analysis of minute shifts from oxidative phosphorylation to anaerobic glycolysis, an early indication of mitochondrial stress. We quantify the dynamics of cellular adaptation to mitochondrial damage and the resulting redistribution of ATP production during rotenone-induced mitochondrial dysfunction and troglitazone (Rezulin)-induced mitochondrial stress. We show troglitazone shifts metabolic fluxes at concentrations previously regarded as safe, suggesting a mechanism for its observed idiosyncratic effect. Our microfluidic platform reveals the dynamics and strategies of cellular adaptation to mitochondrial damage, a unique advantage of organ-on-chip technology.
Collapse
|
165
|
Burkart AM, Tan K, Warren L, Iovino S, Hughes KJ, Kahn CR, Patti ME. Insulin Resistance in Human iPS Cells Reduces Mitochondrial Size and Function. Sci Rep 2016; 6:22788. [PMID: 26948272 PMCID: PMC4780029 DOI: 10.1038/srep22788] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 02/17/2016] [Indexed: 12/20/2022] Open
Abstract
Insulin resistance, a critical component of type 2 diabetes (T2D), precedes and predicts T2D onset. T2D is also associated with mitochondrial dysfunction. To define the cause-effect relationship between insulin resistance and mitochondrial dysfunction, we compared mitochondrial metabolism in induced pluripotent stem cells (iPSC) from 5 healthy individuals and 4 patients with genetic insulin resistance due to insulin receptor mutations. Insulin-resistant iPSC had increased mitochondrial number and decreased mitochondrial size. Mitochondrial oxidative function was impaired, with decreased citrate synthase activity and spare respiratory capacity. Simultaneously, expression of multiple glycolytic enzymes was decreased, while lactate production increased 80%. These perturbations were accompanied by an increase in ADP/ATP ratio and 3-fold increase in AMPK activity, indicating energetic stress. Insulin-resistant iPSC also showed reduced catalase activity and increased susceptibility to oxidative stress. Thus, insulin resistance can lead to mitochondrial dysfunction with reduced mitochondrial size, oxidative activity, and energy production.
Collapse
Affiliation(s)
- Alison M Burkart
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Kelly Tan
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Laura Warren
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Salvatore Iovino
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Katelyn J Hughes
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA, 02215, USA
| | - C Ronald Kahn
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Mary-Elizabeth Patti
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA, 02215, USA
| |
Collapse
|
166
|
Pérez LM, Suárez J, Bernal A, de Lucas B, San Martin N, Gálvez BG. Obesity-driven alterations in adipose-derived stem cells are partially restored by weight loss. Obesity (Silver Spring) 2016; 24:661-9. [PMID: 26833860 DOI: 10.1002/oby.21405] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 10/13/2015] [Accepted: 10/19/2015] [Indexed: 01/03/2023]
Abstract
OBJECTIVE The therapeutic potential of adipose-derived stem cells (ASCs) is reduced by various stress-inducing conditions that affect tissue homeostasis such as diabetes, aging, and obesity. Previous works have provided evidence of negative effects of obesity on ASC populations, but it is unclear whether this persists after a weight loss. This study evaluated whether weight loss can restore the attenuated properties found in ASCs derived from populations with obesity (oASCs). METHODS In vitro functional analyses were performed to investigate the possible recovery properties in mouse oASCs. Using ASCs isolated from subcutaneous tissue from formerly obese mice (dASCs) and control mice (cASCs), cell proliferation, viability, and some regenerative properties in these cells were analyzed compared with oASCs to evaluate the functional cell state. RESULTS Cell proliferation, viability, and some regenerative properties are strengthened in dASCs and cASCs compared with oASCs. Nevertheless, metabolic analysis reveals a mitochondrial load misbalance and function leading to impaired respiration in dASCs. CONCLUSIONS This study demonstrates that an initial obese environment triggers a detrimental state in ASCs that is not completely recovered after weight loss.
Collapse
Affiliation(s)
- Laura M Pérez
- Cardiac Development and Repair Department, Centro Nacional De Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Javier Suárez
- Cardiac Development and Repair Department, Centro Nacional De Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Aurora Bernal
- Cardiac Development and Repair Department, Centro Nacional De Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Beatriz de Lucas
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
| | - Nuria San Martin
- Cardiac Development and Repair Department, Centro Nacional De Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Beatriz G Gálvez
- Cardiac Development and Repair Department, Centro Nacional De Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- School of Doctorate and Research, European University, Madrid, Spain
| |
Collapse
|
167
|
SDF-1/CXCL12 modulates mitochondrial respiration of immature blood cells in a bi-phasic manner. Blood Cells Mol Dis 2016; 58:13-8. [PMID: 27067482 DOI: 10.1016/j.bcmd.2016.01.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 01/26/2016] [Indexed: 01/08/2023]
Abstract
SDF-1/CXCL12 is a potent chemokine required for the homing and engraftment of hematopoietic stem and progenitor cells. Previous data from our group has shown that in an SDF-1/CXCL12 transgenic mouse model, lineage(-) Sca-1(+) c-Kit(+) (LSK) bone marrow cells have reduced mitochondrial membrane potential versus wild-type. These results suggested that SDF-1/CXCL12 may function to keep mitochondrial respiration low in immature blood cells in the bone marrow. Low mitochondrial metabolism helps to maintain low levels of reactive oxygen species (ROS), which can influence differentiation. To test whether SDF-1/CXCL12 regulates mitochondrial metabolism, we employed the human leukemia cell line HL-60, that expresses high levels of the SDF-1/CXCL12 receptor, CXCR4, as a model of hematopoietic progenitor cells in vitro. We treated HL-60 cells with SDF-1/CXCL12 for 2 and 24h. Oxygen consumption rates (OCR), mitochondrial-associated ATP production, mitochondrial mass, and mitochondrial membrane potential of HL-60 cells were significantly reduced at 2h and increased at 24h as compared to untreated control cells. These biphasic effects of SDF-1/CXCL12 were reproduced with lineage negative primary mouse bone marrow cells, suggesting a novel function of SDF-1/CXCL12 in modulating mitochondrial respiration by regulating mitochondrial oxidative phosphorylation, ATP production and mitochondrial content.
Collapse
|
168
|
Falone S, Santini S, di Loreto S, Cordone V, Grannonico M, Cesare P, Cacchio M, Amicarelli F. Improved Mitochondrial and Methylglyoxal-Related Metabolisms Support Hyperproliferation Induced by 50 Hz Magnetic Field in Neuroblastoma Cells. J Cell Physiol 2016; 231:2014-25. [PMID: 26757151 DOI: 10.1002/jcp.25310] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 01/08/2016] [Indexed: 12/27/2022]
Abstract
Extremely low frequency magnetic fields (ELF-MF) are common environmental agents that are suspected to promote later stages of tumorigenesis, especially in brain-derived malignancies. Even though ELF magnetic fields have been previously linked to increased proliferation in neuroblastoma cells, no previous work has studied whether ELF-MF exposure may change key biomolecular features, such as anti-glycative defence and energy re-programming, both of which are currently considered as crucial factors involved in the phenotype and progression of many malignancies. Our study investigated whether the hyperproliferation that is induced in SH-SY5Y human neuroblastoma cells by a 50 Hz, 1 mT ELF magnetic field is supported by an improved defense towards methylglyoxal (MG), which is an endogenous cancer-static and glycating α-oxoaldehyde, and by rewiring of energy metabolism. Our findings show that not only the ELF magnetic field interfered with the biology of neuron-derived malignant cells, by de-differentiating further the cellular phenotype and by increasing the proliferative activity, but also triggered cytoprotective mechanisms through the enhancement of the defense against MG, along with a more efficient management of metabolic energy, presumably to support the rapid cell outgrowth. Intriguingly, we also revealed that the MF-induced bioeffects took place after an initial imbalance of the cellular homeostasis, which most likely created a transient unstable milieu. The biochemical pathways and molecular targets revealed in this research could be exploited for future approaches aimed at limiting or suppressing the deleterious effects of ELF magnetic fields. J. Cell. Physiol. 231: 2014-2025, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Stefano Falone
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Silvano Santini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Silvia di Loreto
- Institute of Translational Pharmacology (IFT)-CNR, L'Aquila, Italy
| | - Valeria Cordone
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Marta Grannonico
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Patrizia Cesare
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Marisa Cacchio
- Department of Biomedical Sciences, University "G. d'Annunzio", Via dei Vestini, Chieti Scalo (CH), Italy
| | - Fernanda Amicarelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.,Institute of Translational Pharmacology (IFT)-CNR, L'Aquila, Italy
| |
Collapse
|
169
|
Schwartz C, Ballinger MA, Andrews MT. Melatonin receptor signaling contributes to neuroprotection upon arousal from torpor in thirteen-lined ground squirrels. Am J Physiol Regul Integr Comp Physiol 2015; 309:R1292-300. [PMID: 26354846 PMCID: PMC4666939 DOI: 10.1152/ajpregu.00292.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/03/2015] [Indexed: 01/13/2023]
Abstract
The brain of mammalian hibernators is naturally protected. Hibernating ground squirrels undergo rapid and extreme changes in body temperature and brain perfusion as they cycle between lengthy torpor bouts and brief periods of euthermia called interbout arousals (IBAs). Arousal from torpor to IBA occurs rapidly, but there is no evidence of brain injury accompanying this extreme physiological transition. Production of the hormone melatonin accompanies arousal, suggesting that it plays a protective role at this time. Here, we investigated mechanisms of melatonin receptor-mediated protection in the brain of the hibernating ground squirrel. We administered the competitive melatonin receptor antagonist luzindole (30 mg/kg ip) to ground squirrels at the predicted end of a torpor bout, triggering an arousal. We found that luzindole-treated animals exhibited caspase-3 activity two times higher than vehicle-treated animals in the hypothalamus at midarousal (P = 0.01), suggesting that melatonin receptor signaling is important for protection in this brain region. We also found a 30% decline in succinate-fueled mitochondrial respiration in luzindole-treated animals compared with vehicle-treated animals (P = 0.019), suggesting that melatonin receptor signaling is important for optimal mitochondrial function during arousal from torpor. The mitochondrial effects of luzindole treatment were seen only during the hibernation season, indicating that this effect is specifically important for arousal from torpor. These data provide evidence for the protective role of melatonin receptor signaling during the extreme physiological transition that occurs when a hibernating mammal arouses from torpor and provide further evidence for regional and seasonal changes in the hibernator brain.
Collapse
MESH Headings
- Adaptation, Physiological
- Animals
- Brain/drug effects
- Brain/metabolism
- Caspase 3/genetics
- Caspase 3/metabolism
- Central Nervous System/physiology
- Female
- Gene Expression Regulation, Enzymologic
- Hibernation/drug effects
- Hibernation/physiology
- Male
- Melatonin/metabolism
- Mitochondria/drug effects
- Mitochondria/metabolism
- Receptor, Melatonin, MT1/antagonists & inhibitors
- Receptor, Melatonin, MT1/metabolism
- Receptor, Melatonin, MT2/antagonists & inhibitors
- Receptor, Melatonin, MT2/metabolism
- Sciuridae/physiology
- Seasons
- Signal Transduction/physiology
- Tryptamines/pharmacology
Collapse
Affiliation(s)
- Christine Schwartz
- Department of Biology, University of Minnesota Duluth, Duluth, Minnesota; and Department of Biology, University of Wisconsin-La Crosse, La Crosse, Wisconsin
| | - Mallory A Ballinger
- Department of Biology, University of Minnesota Duluth, Duluth, Minnesota; and
| | - Matthew T Andrews
- Department of Biology, University of Minnesota Duluth, Duluth, Minnesota; and
| |
Collapse
|
170
|
McAninch EA, Miller BT, Ueta CB, Jo S, Kim BW. Thyroid Hormone at Near Physiologic Concentrations Acutely Increases Oxygen Consumption and Extracellular Acidification in LH86 Hepatoma Cells. Endocrinology 2015; 156:4325-35. [PMID: 26287403 DOI: 10.1210/en.2015-1221] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Thyroid hormone (T3) has been known to regulate the basal metabolic rate for more than a century, but mechanistic understanding is lacking both at the level of the intact organism and in terms of how T3 alters energy expenditure in individual tissues. The current studies investigate the question of which metabolically relevant genes respond acutely as T3 concentrations increase through the physiologic range in liver cells. Because this has been technically unfeasible historically, we developed a modified protocol for extracellular flux analysis using a 96-well Extracellular Flux Analyzer (Seahorse Bioscience). Using a modified extracellular flux protocol and LH86 human hepatoma cells, we established an experimental system where small but significant changes in O2 consumption could be reproducibly quantified as hypothyroid cells were exposed to near-physiologic final concentrations of T3 approximately 2 orders of magnitude lower than most studies (0.04 nM free T3), in only 6-7 hours. Taking advantage of the nondestructive nature of 96-well Extracellular Flux Analyzer measurements, the acute, direct, transcriptional changes that occur were measured in the exact same cells demonstrating increased O2 consumption. An unbiased, genome-wide microarray analysis identified potential candidate genes related to fatty acid oxidation, angiogenesis, nucleotide metabolism, immune signaling, mitochondrial respiration, and cell proliferation. The identified transcriptome is likely enriched in the genes most important for mediating the energetic effects of T3 in hepatoma cells.
Collapse
Affiliation(s)
- Elizabeth A McAninch
- Division of Endocrinology and Metabolism (E.A.M., S.J., B.W.K.), Rush University Medical Center, Chicago, Illinois 60612; University of South Florida Morsani College of Medicine (B.T.M.), Tampa, Florida 33612; and Institute of Biomedical Science (C.B.U.), University of São Paulo, São Paulo SP 05508-000, Brazil
| | - Bradford T Miller
- Division of Endocrinology and Metabolism (E.A.M., S.J., B.W.K.), Rush University Medical Center, Chicago, Illinois 60612; University of South Florida Morsani College of Medicine (B.T.M.), Tampa, Florida 33612; and Institute of Biomedical Science (C.B.U.), University of São Paulo, São Paulo SP 05508-000, Brazil
| | - Cintia B Ueta
- Division of Endocrinology and Metabolism (E.A.M., S.J., B.W.K.), Rush University Medical Center, Chicago, Illinois 60612; University of South Florida Morsani College of Medicine (B.T.M.), Tampa, Florida 33612; and Institute of Biomedical Science (C.B.U.), University of São Paulo, São Paulo SP 05508-000, Brazil
| | - Sungro Jo
- Division of Endocrinology and Metabolism (E.A.M., S.J., B.W.K.), Rush University Medical Center, Chicago, Illinois 60612; University of South Florida Morsani College of Medicine (B.T.M.), Tampa, Florida 33612; and Institute of Biomedical Science (C.B.U.), University of São Paulo, São Paulo SP 05508-000, Brazil
| | - Brian W Kim
- Division of Endocrinology and Metabolism (E.A.M., S.J., B.W.K.), Rush University Medical Center, Chicago, Illinois 60612; University of South Florida Morsani College of Medicine (B.T.M.), Tampa, Florida 33612; and Institute of Biomedical Science (C.B.U.), University of São Paulo, São Paulo SP 05508-000, Brazil
| |
Collapse
|
171
|
Anthony Jalin AMA, Rajasekaran M, Prather PL, Kwon JS, Gajulapati V, Choi Y, Kim C, Pahk K, Ju C, Kim WK. Non-Selective Cannabinoid Receptor Antagonists, Hinokiresinols Reduce Infiltration of Microglia/Macrophages into Ischemic Brain Lesions in Rat via Modulating 2-Arachidonolyglycerol-Induced Migration and Mitochondrial Activity. PLoS One 2015; 10:e0141600. [PMID: 26517721 PMCID: PMC4627794 DOI: 10.1371/journal.pone.0141600] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 10/09/2015] [Indexed: 11/19/2022] Open
Abstract
Growing evidence suggests that therapeutic strategies to modulate the post-ischemic inflammatory responses are promising approaches to improve stroke outcome. Although the endocannabinoid system has been emerged as an endogenous therapeutic target to regulate inflammation after stroke insult, the downstream mechanisms and their potentials for therapeutic intervention remain controversial. Here we identified trans- and cis-hinokiresinols as novel non-selective antagonists for two G-protein-coupled cannabinoid receptors, cannabinoid receptor type 1 and type 2. The Electric Cell-substrate Impedance Sensing and Boyden chamber migration assays using primary microglial cultures revealed that both hinokiresinols significantly inhibited an endocannabinoid, 2-arachidonoylglycerol-induced migration. Hinokiresinols modulated 2-arachidonoylglycerol-induced mitochondrial bioenergetics in microglia as evidenced by inhibition of ATP turnover and reduction in respiratory capacity, thereby resulting in impaired migration activity. In rats subjected to transient middle cerebral artery occlusion (1.5-h) followed by 24-h reperfusion, post-ischemic treatment with hinokiresinols (2 and 7-h after the onset of ischemia, 10 mg/kg) significantly reduced cerebral infarct and infiltration of ED1-positive microglial/macrophage cells into cerebral ischemic lesions in vivo. Co-administration of exogenous 2-AG (1 mg/kg, i.v., single dose at 2 h after starting MCAO) abolished the protective effect of trans-hinokiresionol. These results suggest that hinokiresinols may serve as stroke treatment by targeting the endocannabinoid system. Alteration of mitochondrial bioenergetics and consequent inhibition of inflammatory cells migration may be a novel mechanism underlying anti-ischemic effects conferred by cannabinoid receptor antagonists.
Collapse
Affiliation(s)
| | - Maheswari Rajasekaran
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Paul L. Prather
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Jin Sun Kwon
- Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Veeraswamy Gajulapati
- Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Yongseok Choi
- Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Chunsook Kim
- Department of Nursing, Kyungdong University, Wonju, Kangwon-do, Republic of Korea
| | - Kisoo Pahk
- Department of Neuroscience, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Chung Ju
- Department of Neuroscience, College of Medicine, Korea University, Seoul, Republic of Korea
- * E-mail: (W-KK); (CJ)
| | - Won-Ki Kim
- Department of Neuroscience, College of Medicine, Korea University, Seoul, Republic of Korea
- * E-mail: (W-KK); (CJ)
| |
Collapse
|
172
|
Zhang J, Wang C, Chen X, Takada M, Fan C, Zheng X, Wen H, Liu Y, Wang C, Pestell RG, Aird KM, Kaelin WG, Liu XS, Zhang Q. EglN2 associates with the NRF1-PGC1α complex and controls mitochondrial function in breast cancer. EMBO J 2015; 34:2953-70. [PMID: 26492917 DOI: 10.15252/embj.201591437] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 09/11/2015] [Indexed: 12/15/2022] Open
Abstract
The EglN2/PHD1 prolyl hydroxylase is an important oxygen sensor contributing to breast tumorigenesis. Emerging studies suggest that there is functional cross talk between oxygen sensing and mitochondrial function, both of which play an essential role for sustained tumor growth. However, the potential link between EglN2 and mitochondrial function remains largely undefined. Here, we show that EglN2 depletion decreases mitochondrial respiration in breast cancer under normoxia and hypoxia, which correlates with decreased mitochondrial DNA in a HIF1/2α-independent manner. Integrative analyses of gene expression profile and genomewide binding of EglN2 under hypoxic conditions reveal nuclear respiratory factor 1 (NRF1) motif enrichment in EglN2-activated genes, suggesting NRF1 as an EglN2 binding partner. Mechanistically, by forming an activator complex with PGC1α and NRF1 on chromatin, EglN2 promotes the transcription of ferridoxin reductase (FDXR) and maintains mitochondrial function. In addition, FDXR, as one of effectors for EglN2, contributes to breast tumorigenesis in vitro and in vivo. Our findings suggest that EglN2 regulates mitochondrial function in ERα-positive breast cancer.
Collapse
Affiliation(s)
- Jing Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Chengyang Wang
- Department of Bioinformatics, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Xi Chen
- Department of Molecular and Cellular Biology, The Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Mamoru Takada
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Cheng Fan
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Xingnan Zheng
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Haitao Wen
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA Department of Surgery, University of North Carolina, Chapel Hill, NC, USA
| | - Yong Liu
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Chenguang Wang
- Program of Radiation Protection and Drug Discovery, Institute of Radiation Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin, China
| | - Richard G Pestell
- Department of Cancer Biology and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Katherine M Aird
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA, USA
| | - William G Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Xiaole Shirley Liu
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard School of Public Health, Boston, MA, USA
| | - Qing Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
173
|
Cardaci S, Zheng L, MacKay G, van den Broek NJ, MacKenzie ED, Nixon C, Stevenson D, Tumanov S, Bulusu V, Kamphorst JJ, Vazquez A, Fleming S, Schiavi F, Kalna G, Blyth K, Strathdee D, Gottlieb E. Pyruvate carboxylation enables growth of SDH-deficient cells by supporting aspartate biosynthesis. Nat Cell Biol 2015; 17:1317-26. [PMID: 26302408 PMCID: PMC4591470 DOI: 10.1038/ncb3233] [Citation(s) in RCA: 216] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 07/29/2015] [Indexed: 12/13/2022]
Abstract
Succinate dehydrogenase (SDH) is a heterotetrameric nuclear-encoded complex responsible for the oxidation of succinate to fumarate in the tricarboxylic acid cycle. Loss-of-function mutations in any of the SDH genes are associated with cancer formation. However, the impact of SDH loss on cell metabolism and the mechanisms enabling growth of SDH-defective cells are largely unknown. Here, we generated Sdhb-ablated kidney mouse cells and used comparative metabolomics and stable-isotope-labelling approaches to identify nutritional requirements and metabolic adaptations to SDH loss. We found that lack of SDH activity commits cells to consume extracellular pyruvate, which sustains Warburg-like bioenergetic features. We further demonstrated that pyruvate carboxylation diverts glucose-derived carbons into aspartate biosynthesis, thus sustaining cell growth. By identifying pyruvate carboxylase as essential for the proliferation and tumorigenic capacity of SDH-deficient cells, this study revealed a metabolic vulnerability for potential future treatment of SDH-associated malignancies.
Collapse
MESH Headings
- Animals
- Aspartic Acid/biosynthesis
- Carboxylic Acids/metabolism
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Cell Line, Transformed
- Cell Proliferation
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cells, Cultured
- Humans
- Immunoblotting
- Kidney/cytology
- Kidney/metabolism
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Male
- Metabolomics/methods
- Mice, 129 Strain
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Nude
- Pyruvate Carboxylase/metabolism
- Pyruvic Acid/metabolism
- RNA Interference
- Succinate Dehydrogenase/genetics
- Succinate Dehydrogenase/metabolism
Collapse
Affiliation(s)
- Simone Cardaci
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| | - Liang Zheng
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| | - Gillian MacKay
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| | | | | | - Colin Nixon
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| | - David Stevenson
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| | - Sergey Tumanov
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1BD, UK
| | - Vinay Bulusu
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1BD, UK
| | - Jurre J. Kamphorst
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1BD, UK
| | - Alexei Vazquez
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| | - Stewart Fleming
- Department of Pathology, University of Dundee, Ninewells Hospital, Dundee, DD1 9SY, UK
| | - Francesca Schiavi
- Veneto Institute of Oncology IRCCS, Familial cancer clinic and oncoendocrinology, Via Gattamelata 64, 35128 Padova, Italy
| | - Gabriela Kalna
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| | - Karen Blyth
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| | - Douglas Strathdee
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| | - Eyal Gottlieb
- Cancer Research UK, Beatson Institute, Switchback Rd, Glasgow, G61 1BD, UK
| |
Collapse
|
174
|
Perspectives on the membrane fatty acid unsaturation/pacemaker hypotheses of metabolism and aging. Chem Phys Lipids 2015; 191:48-60. [PMID: 26291495 DOI: 10.1016/j.chemphyslip.2015.08.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/10/2015] [Accepted: 08/12/2015] [Indexed: 12/27/2022]
Abstract
The membrane pacemaker hypotheses of metabolism and aging are distinct, but interrelated hypotheses positing that increases in unsaturation of lipids within membranes are correlated with increasing basal metabolic rate and decreasing longevity, respectively. The two hypotheses each have evidence that either supports or contradicts them, but consensus has failed to emerge. In this review, we identify sources of weakness of previous studies supporting and contradicting these hypotheses and suggest different methods and lines of inquiry. The link between fatty acyl composition of membranes and membrane-bound protein activity is a central tenet of the membrane pacemaker hypothesis of metabolism, but the mechanism by which unsaturation would change protein activity is not well defined and, whereas fatty acid desaturases have been put forward by some as the mechanism behind evolutionary differences in fatty acyl composition of phospholipids among organisms, there have been no studies to differentiate whether desaturases have been more affected by natural selection on aging and metabolic rate than have elongases or acyltransferases. Past analyses have been hampered by potentially incorrect estimates of the peroxidizability of lipids and longevity of study animals, and by the confounding effect of phylogeny. According to some authors, body mass may also be a confounding effect that should be taken into account, though this is not universally accepted. Further research on this subject should focus more on mechanisms and take weaknesses of past studies into account.
Collapse
|
175
|
Takebayashi SI, Tanaka H, Hino S, Nakatsu Y, Igata T, Sakamoto A, Narita M, Nakao M. Retinoblastoma protein promotes oxidative phosphorylation through upregulation of glycolytic genes in oncogene-induced senescent cells. Aging Cell 2015; 14:689-97. [PMID: 26009982 PMCID: PMC4531082 DOI: 10.1111/acel.12351] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2015] [Indexed: 12/13/2022] Open
Abstract
Metabolism is closely linked with cellular state and biological processes, but the mechanisms controlling metabolic properties in different contexts remain unclear. Cellular senescence is an irreversible growth arrest induced by various stresses, which exhibits active secretory and metabolic phenotypes. Here, we show that retinoblastoma protein (RB) plays a critical role in promoting the metabolic flow by activating both glycolysis and mitochondrial oxidative phosphorylation (OXPHOS) in cells that have undergone oncogene-induced senescence (OIS). A combination of real-time metabolic monitoring, and metabolome and gene expression analyses showed that OIS-induced fibroblasts developed an accelerated metabolic flow. The loss of RB downregulated a series of glycolytic genes and simultaneously reduced metabolites produced from the glycolytic pathway, indicating that RB upregulates glycolytic genes in OIS cells. Importantly, both mitochondrial OXPHOS and glycolytic activities were abolished in RB-depleted or downstream glycolytic enzyme-depleted OIS cells, suggesting that RB-mediated glycolytic activation induces a metabolic flux into the OXPHOS pathway. Collectively, our findings reveal that RB essentially functions in metabolic remodeling and the maintenance of the active energy production in OIS cells.
Collapse
Affiliation(s)
- Shin-ichiro Takebayashi
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamoto, Japan
- Program for Leading Graduate Schools ‘HIGO (Health life science: Interdisciplinary and Glocal Oriented) Program’, Kumamoto UniversityKumamoto, Japan
| | - Hiroshi Tanaka
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamoto, Japan
| | - Shinjiro Hino
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamoto, Japan
| | - Yuko Nakatsu
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamoto, Japan
| | - Tomoka Igata
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamoto, Japan
| | - Akihisa Sakamoto
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamoto, Japan
| | - Masashi Narita
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridge, UK
| | - Mitsuyoshi Nakao
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto UniversityKumamoto, Japan
- Program for Leading Graduate Schools ‘HIGO (Health life science: Interdisciplinary and Glocal Oriented) Program’, Kumamoto UniversityKumamoto, Japan
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology AgencyTokyo, Japan
| |
Collapse
|
176
|
Chen CNJ, Lin SY, Liao YH, Li ZJ, Wong AMK. Late-onset caloric restriction alters skeletal muscle metabolism by modulating pyruvate metabolism. Am J Physiol Endocrinol Metab 2015; 308:E942-9. [PMID: 26032513 DOI: 10.1152/ajpendo.00508.2014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 04/07/2015] [Indexed: 12/20/2022]
Abstract
Caloric restriction (CR) attenuates age-related muscle loss. However, the underlying mechanism responsible for this attenuation is not fully understood. This study evaluated the role of energy metabolism in the CR-induced attenuation of muscle loss. The aims of this study were twofold: 1) to evaluate the effect of CR on energy metabolism and determine its relationship with muscle mass, and 2) to determine whether the effects of CR are age dependent. Young and middle-aged rats were randomized into either 40% CR or ad libitum (AL) diet groups for 14 wk. Major energy-producing pathways in muscles, i.e., glycolysis and mitochondrial oxidative phosphorylation (OXPHOS), were examined. We found that the effects of CR were age dependent. CR improved muscle metabolism and normalized muscle mass in middle-aged animals but not young animals. CR decreased glycolysis and increased the cellular dependency for OXPHOS vs. glycolysis in muscles of middle-aged rats, which was associated with the improvement of normalized muscle mass. The metabolic reprogramming induced by CR was related to modulation of pyruvate metabolism and increased mitochondrial biogenesis. Compared with animals fed AL, middle-aged animals with CR had lower lactate dehydrogenase A content and greater mitochondrial pyruvate carrier content. Markers of mitochondrial biogenesis, including AMPK activation levels and SIRT1 and COX-IV content, also showed increased levels. In conclusion, 14 wk of CR improved muscle metabolism and preserved muscle mass in middle-aged animals but not in young developing animals. CR-attenuated age-related muscle loss is associated with reprogramming of the metabolic pathway from glycolysis to OXPHOS.
Collapse
Affiliation(s)
- Chiao-Nan Joyce Chen
- Department of Physical Therapy, Medical School, Chang Gung University, Tao-Yuan, Taiwan; Healthy Aging Research Center, Chang Gung University, Tao-Yuan, Taiwan;
| | - Shang-Ying Lin
- Department of Physical Therapy, Medical School, Chang Gung University, Tao-Yuan, Taiwan
| | - Yi-Hung Liao
- Department of Exercise and Health Science, College of Human Development and Health, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan; and
| | - Zhen-Jie Li
- Department of Physical Therapy, Medical School, Chang Gung University, Tao-Yuan, Taiwan
| | - Alice May-Kuen Wong
- Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
177
|
The metabolic rate of cultured muscle cells from hybrid Coturnix quail is intermediate to that of muscle cells from fast-growing and slow-growing Coturnix quail. J Comp Physiol B 2015; 185:547-57. [DOI: 10.1007/s00360-015-0906-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 04/15/2015] [Accepted: 04/26/2015] [Indexed: 10/23/2022]
|
178
|
Pérez LM, Bernal A, de Lucas B, San Martin N, Mastrangelo A, García A, Barbas C, Gálvez BG. Altered metabolic and stemness capacity of adipose tissue-derived stem cells from obese mouse and human. PLoS One 2015; 10:e0123397. [PMID: 25875023 PMCID: PMC4395137 DOI: 10.1371/journal.pone.0123397] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 02/18/2015] [Indexed: 12/21/2022] Open
Abstract
Adipose stem cells (ASCs) are an appealing source of cells for therapeutic intervention; however, the environment from which ASCs are isolated may impact their usefulness. Using a range of functional assays, we have evaluated whether ASCs isolated from an obese environment are comparable to cells from non-obese adipose tissue. Results showed that ASCs isolated from obese tissue have a reduced proliferative ability and a loss of viability together with changes in telomerase activity and DNA telomere length, suggesting a decreased self-renewal capacity. Metabolic analysis demonstrated that mitochondrial content and function was impaired in obese-derived ASCs resulting in changes in favored oxidative substrates. These findings highlight the impact of obesity on adult stem properties. Hence, caution should be exercised when considering the source of ASCs for cellular therapies since their therapeutic potential may be impaired.
Collapse
Affiliation(s)
- Laura M. Pérez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Aurora Bernal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Beatriz de Lucas
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Nuria San Martin
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | | | | | - Beatriz G. Gálvez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Universidad Europea de Madrid (UEM), Madrid, Spain
- * E-mail:
| |
Collapse
|
179
|
Kropp EM, Oleson BJ, Broniowska KA, Bhattacharya S, Chadwick AC, Diers AR, Hu Q, Sahoo D, Hogg N, Boheler KR, Corbett JA, Gundry RL. Inhibition of an NAD⁺ salvage pathway provides efficient and selective toxicity to human pluripotent stem cells. Stem Cells Transl Med 2015; 4:483-93. [PMID: 25834119 DOI: 10.5966/sctm.2014-0163] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 02/16/2015] [Indexed: 11/16/2022] Open
Abstract
The tumorigenic potential of human pluripotent stem cells (hPSCs) is a major limitation to the widespread use of hPSC derivatives in the clinic. Here, we demonstrate that the small molecule STF-31 is effective at eliminating undifferentiated hPSCs across a broad range of cell culture conditions with important advantages over previously described methods that target metabolic processes. Although STF-31 was originally described as an inhibitor of glucose transporter 1, these data support the reclassification of STF-31 as a specific NAD⁺ salvage pathway inhibitor through the inhibition of nicotinamide phosphoribosyltransferase (NAMPT). These findings demonstrate the importance of an NAD⁺ salvage pathway in hPSC biology and describe how inhibition of NAMPT can effectively eliminate hPSCs from culture. These results will advance and accelerate the development of safe, clinically relevant hPSC-derived cell-based therapies.
Collapse
Affiliation(s)
- Erin M Kropp
- Department of Biochemistry, Department of Biophysics, Redox Biology Program, and Department of Medicine, Division of Endocrinology, Metabolism and Clinical Nutrition, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Department of Physiology, Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong, Special Administrative Region of the People's Republic of China; Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Bryndon J Oleson
- Department of Biochemistry, Department of Biophysics, Redox Biology Program, and Department of Medicine, Division of Endocrinology, Metabolism and Clinical Nutrition, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Department of Physiology, Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong, Special Administrative Region of the People's Republic of China; Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Katarzyna A Broniowska
- Department of Biochemistry, Department of Biophysics, Redox Biology Program, and Department of Medicine, Division of Endocrinology, Metabolism and Clinical Nutrition, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Department of Physiology, Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong, Special Administrative Region of the People's Republic of China; Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Subarna Bhattacharya
- Department of Biochemistry, Department of Biophysics, Redox Biology Program, and Department of Medicine, Division of Endocrinology, Metabolism and Clinical Nutrition, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Department of Physiology, Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong, Special Administrative Region of the People's Republic of China; Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alexandra C Chadwick
- Department of Biochemistry, Department of Biophysics, Redox Biology Program, and Department of Medicine, Division of Endocrinology, Metabolism and Clinical Nutrition, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Department of Physiology, Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong, Special Administrative Region of the People's Republic of China; Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anne R Diers
- Department of Biochemistry, Department of Biophysics, Redox Biology Program, and Department of Medicine, Division of Endocrinology, Metabolism and Clinical Nutrition, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Department of Physiology, Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong, Special Administrative Region of the People's Republic of China; Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Qinghui Hu
- Department of Biochemistry, Department of Biophysics, Redox Biology Program, and Department of Medicine, Division of Endocrinology, Metabolism and Clinical Nutrition, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Department of Physiology, Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong, Special Administrative Region of the People's Republic of China; Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daisy Sahoo
- Department of Biochemistry, Department of Biophysics, Redox Biology Program, and Department of Medicine, Division of Endocrinology, Metabolism and Clinical Nutrition, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Department of Physiology, Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong, Special Administrative Region of the People's Republic of China; Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Neil Hogg
- Department of Biochemistry, Department of Biophysics, Redox Biology Program, and Department of Medicine, Division of Endocrinology, Metabolism and Clinical Nutrition, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Department of Physiology, Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong, Special Administrative Region of the People's Republic of China; Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kenneth R Boheler
- Department of Biochemistry, Department of Biophysics, Redox Biology Program, and Department of Medicine, Division of Endocrinology, Metabolism and Clinical Nutrition, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Department of Physiology, Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong, Special Administrative Region of the People's Republic of China; Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John A Corbett
- Department of Biochemistry, Department of Biophysics, Redox Biology Program, and Department of Medicine, Division of Endocrinology, Metabolism and Clinical Nutrition, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Department of Physiology, Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong, Special Administrative Region of the People's Republic of China; Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rebekah L Gundry
- Department of Biochemistry, Department of Biophysics, Redox Biology Program, and Department of Medicine, Division of Endocrinology, Metabolism and Clinical Nutrition, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Department of Physiology, Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong, Special Administrative Region of the People's Republic of China; Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
180
|
Dutta A, Chan SHP, Pauli NT, Raina R. Hypersensitive response-like lesions 1 codes for AtPPT1 and regulates accumulation of ROS and defense against bacterial pathogen Pseudomonas syringae in Arabidopsis thaliana. Antioxid Redox Signal 2015; 22:785-96. [PMID: 25557512 PMCID: PMC4361009 DOI: 10.1089/ars.2014.5963] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
AIMS Plants employ both basal and resistance gene (R gene)-mediated defenses in response to pathogens. Reactive oxygen species (ROS) are widely reported to play a central role in both basal and R gene-mediated defense; however, the nature of ROS has been less well established for basal defense. In addition, spatial distribution of redox moieties and mechanisms of plant responses during basal defense are poorly understood. We investigated redox signaling in Arabidopsis thaliana in response to virulent bacterial pathogen, focusing on the role of the mitochondria in balancing energy demands against generation of physiologically relevant ROS. RESULTS Positional cloning of an Arabidopsis lesion mimic mutant identified a polyprenyl transferase involved in the biosynthesis of Coenzyme Q10 (CoQ), which leads to novel insights into physiological ROS levels and their role in basal resistance. Gain- and loss-of-function studies identified Coenzyme Q10 redox state to be a key determinant of ROS levels. These Coenzyme Q10 redox state-mediated ROS levels had a direct bearing on both response against pathogen and ability to thrive in high oxidative stress environments. INNOVATION We demonstrate that Coenzyme Q10 redox state generates an ROS threshold for a successful basal resistance response. Perturbation of the Coenzyme Q10 redox state has the potential to disrupt plant defense responses against bacterial pathogens. CONCLUSIONS Coenzyme Q10 redox state is a key regulator of Arabidopsis basal resistance against bacterial pathogens.
Collapse
Affiliation(s)
- Aditya Dutta
- Department of Biology, Syracuse University , Syracuse, New York
| | | | | | | |
Collapse
|
181
|
Lin HY, Liou CW, Chen SD, Hsu TY, Chuang JH, Wang PW, Huang ST, Tiao MM, Chen JB, Lin TK, Chuang YC. Mitochondrial transfer from Wharton's jelly-derived mesenchymal stem cells to mitochondria-defective cells recaptures impaired mitochondrial function. Mitochondrion 2015; 22:31-44. [PMID: 25746175 DOI: 10.1016/j.mito.2015.02.006] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 02/22/2015] [Accepted: 02/24/2015] [Indexed: 02/01/2023]
Abstract
Adult mesenchymal stem cell (MSC)-conducted mitochondrial transfer has been recently shown to rescue cellular bioenergetics and prevent cell death caused by mitochondrial dysfunction. Wharton's jelly-derived MSCs (WJMSCs) harvested from postpartum umbilical cords are an accessible and abundant source of stem cells. This study aimed to determine the capability of WJMSCs to transfer their own mitochondria and rescue impaired oxidative phosphorylation (OXPHOS) and bioenergetics caused by mitochondrial DNA defects. To do this, WJMSCs were co-cultured with mitochondrial DNA (mtDNA)-depleted ρ(0) cells and the recapture of mitochondrial function was evaluated. WJMSCs were shown to be capable of transferring their own mitochondria into ρ(0) cells and underwent interorganellar mixture within these cells. Permissive culture media (BrdU-containing and pyruvate- and uridine-free) sieved out a survival cell population from the co-cultured WJMSCs (BrdU-sensitive) and ρ(0) cells (pyruvate/uridine-free). The survival cells had mtDNA identical to that of WJMSCs, whereas they expressed cellular markers identical to that of ρ(0) cells. Importantly, these ρ(0)-plus -WJMSC-mtDNA (ρ(+W)) cells recovered the expression of mtDNA-encoded proteins and exhibited functional oxygen consumption and respiratory control, as well as the activity of electron transport chain (ETC) complexes I, II, III and IV. In addition, ETC complex V-inhibitor-sensitive ATP production and metabolic shifting were also recovered. Furthermore, cellular behaviors including attachment-free proliferation, aerobic viability and OXPHOS-reliant cellular motility were also regained after mitochondrial transfer by WJMSCs. The therapeutic effect of WJMSCs-derived mitochondrial transfer was able to stably sustain for at least 45 passages. In conclusion, this study suggests that WJMSCs may serve as a potential therapeutic strategy for diseases linked to mitochondrial dysfunction through the donation of healthy mitochondria to cells with genetic mitochondrial defects.
Collapse
Affiliation(s)
- Hung-Yu Lin
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; Mitochondrial Research Unit, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; Center of Parkinson's Disease, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
| | - Chia-Wei Liou
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; Mitochondrial Research Unit, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; Center of Parkinson's Disease, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Shang-Der Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; Mitochondrial Research Unit, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; Center of Parkinson's Disease, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
| | - Te-Yao Hsu
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Jiin-Haur Chuang
- Mitochondrial Research Unit, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; Division of Pediatric Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Pei-Wen Wang
- Mitochondrial Research Unit, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Sheng-Teng Huang
- Mitochondrial Research Unit, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; Department of Chinese Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Mao-Meng Tiao
- Mitochondrial Research Unit, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Jin-Bor Chen
- Mitochondrial Research Unit, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; Department of Nephrology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Tsu-Kung Lin
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; Mitochondrial Research Unit, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; Center of Parkinson's Disease, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Yao-Chung Chuang
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; Center of Parkinson's Disease, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
182
|
Kao YT, Chang BL, Liang JJ, Tsai HJ, Lee YL, Lin RJ, Lin YL. Japanese encephalitis virus nonstructural protein NS5 interacts with mitochondrial trifunctional protein and impairs fatty acid β-oxidation. PLoS Pathog 2015; 11:e1004750. [PMID: 25816318 PMCID: PMC4376648 DOI: 10.1371/journal.ppat.1004750] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 02/17/2015] [Indexed: 01/16/2023] Open
Abstract
Infection with Japanese encephalitis virus (JEV) can induce the expression of pro-inflammatory cytokines and cause acute encephalitis in humans. β-oxidation breaks down fatty acids for ATP production in mitochondria, and impaired β-oxidation can induce pro-inflammatory cytokine expression. To address the role of fatty-acid β-oxidation in JEV infection, we measured the oxygen consumption rate of mock- and JEV-infected cells cultured with or without long chain fatty acid (LCFA) palmitate. Cells with JEV infection showed impaired LCFA β-oxidation and increased interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α) expression. JEV nonstructural protein 5 (NS5) interacted with hydroxyacyl-CoA dehydrogenase α and β subunits, two components of the mitochondrial trifunctional protein (MTP) involved in LCFA β-oxidation, and NS5 proteins were detected in mitochondria and co-localized with MTP. LCFA β-oxidation was impaired and higher cytokines were induced in cells overexpressing NS5 protein as compared with control cells. Deletion and mutation studies showed that the N-terminus of NS5 was involved in the MTP association, and a single point mutation of NS5 residue 19 from methionine to alanine (NS5-M19A) reduced its binding ability with MTP. The recombinant JEV with NS5-M19A mutation (JEV-NS5-M19A) was less able to block LCFA β-oxidation and induced lower levels of IL-6 and TNF-α than wild-type JEV. Moreover, mice challenged with JEV-NS5-M19A showed less neurovirulence and neuroinvasiveness. We identified a novel function of JEV NS5 in viral pathogenesis by impairing LCFA β-oxidation and inducing cytokine expression by association with MTP.
Collapse
Affiliation(s)
- Yu-Ting Kao
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Bi-Lan Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jian-Jong Liang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hang-Jen Tsai
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Ling Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ren-Jye Lin
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Primary Care Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yi-Ling Lin
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
183
|
Baker CN, Gidus SA, Price GF, Peoples JNR, Ebert SN. Impaired cardiac energy metabolism in embryos lacking adrenergic stimulation. Am J Physiol Endocrinol Metab 2015; 308:E402-13. [PMID: 25516547 PMCID: PMC4346738 DOI: 10.1152/ajpendo.00267.2014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
As development proceeds from the embryonic to fetal stages, cardiac energy demands increase substantially, and oxidative phosphorylation of ADP to ATP in mitochondria becomes vital. Relatively little, however, is known about the signaling mechanisms regulating the transition from anaerobic to aerobic metabolism that occurs during the embryonic period. The main objective of this study was to test the hypothesis that adrenergic hormones provide critical stimulation of energy metabolism during embryonic/fetal development. We examined ATP and ADP concentrations in mouse embryos lacking adrenergic hormones due to targeted disruption of the essential dopamine β-hydroxylase (Dbh) gene. Embryonic ATP concentrations decreased dramatically, whereas ADP concentrations rose such that the ATP/ADP ratio in the adrenergic-deficient group was nearly 50-fold less than that found in littermate controls by embryonic day 11.5. We also found that cardiac extracellular acidification and oxygen consumption rates were significantly decreased, and mitochondria were significantly larger and more branched in adrenergic-deficient hearts. Notably, however, the mitochondria were intact with well-formed cristae, and there was no significant difference observed in mitochondrial membrane potential. Maternal administration of the adrenergic receptor agonists isoproterenol or l-phenylephrine significantly ameliorated the decreases in ATP observed in Dbh-/- embryos, suggesting that α- and β-adrenergic receptors were effective modulators of ATP concentrations in mouse embryos in vivo. These data demonstrate that adrenergic hormones stimulate cardiac energy metabolism during a critical period of embryonic development.
Collapse
Affiliation(s)
- Candice N Baker
- Burnett School of Biomedical Sciences, University of Central Florida, College of Medicine, Orlando, Florida; and
| | - Sarah A Gidus
- Burnett School of Biomedical Sciences, University of Central Florida, College of Medicine, Orlando, Florida; and
| | - George F Price
- Department of Electron Microscopy, Department of Pathology, Orlando Regional Medical Center, Orlando, Florida
| | - Jessica N R Peoples
- Burnett School of Biomedical Sciences, University of Central Florida, College of Medicine, Orlando, Florida; and
| | - Steven N Ebert
- Burnett School of Biomedical Sciences, University of Central Florida, College of Medicine, Orlando, Florida; and
| |
Collapse
|
184
|
Wang DF, Rong WT, Lu Y, Hou J, Qi SS, Xiao Q, Zhang J, You J, Yu SQ, Xu Q. TPGS2k/PLGA nanoparticles for overcoming multidrug resistance by interfering mitochondria of human alveolar adenocarcinoma cells. ACS APPLIED MATERIALS & INTERFACES 2015; 7:3888-3901. [PMID: 25644220 DOI: 10.1021/am508340m] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
In this study, we successfully synthesized d-α-tocopheryl polyethylene glycol 2000 succinate (TPGS2k) and prepared TPGS2k-modified poly(lactic-co-glycolic acid) nanoparticles (TPGS2k/PLGA NPs) loaded with 7-ethyl-10-hydroxycamptothecin (SN-38), designated TPGS2k/PLGA/SN-38 NPs. Characterization measurements showed that TPGS2k/PLGA/SN-38 NPs displayed flat and spheroidal particles with diameters of 80-104 nm. SN-38 was encapsulated in TPGS2k emulsified PLGA NPs with the entrapment efficiency and loading rates of SN-38 83.6 and 7.85%, respectively. SN-38 could release constantly from TPGS2k/PLGA/SN-38 NPs in vitro. TPGS2k/PLGA/SN-38 NPs induced significantly higher cytotoxicity on A549 cells and the multidrug resistance (MDR) cell line (A549/DDP cells and A549/Taxol cells) compared with free SN-38. Further studies on the mechanism of the NPs in increasing the death of MDR cells showed that following the SN-38 releasing into cytoplasm the remaining TPGS2k/PLGA NPs could reverse the P-gp mediated MDR via interfering with the structure and function of mitochondria and rather than directly inhibiting the enzymatic activity of P-gp ATPase. Therefore, TPGS2k/PLGA NPs can reduce the generation of ATP and the release of energy for the requisite of P-gp efflux transporters. The results indicated that TPGS2k/PLGA NPs could become the nanopharmaceutical materials with the capability to reversal MDR and improve anticancer effects of some chemotherapy drugs as P-gp substrates.
Collapse
Affiliation(s)
- Dong-Fang Wang
- Jiangsu Key Laboratory for Supramolecular Medicinal Materials and Applications, College of Life Sciences, Nanjing Normal University , Nanjing 210046, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Krajcova A, Ziak J, Jiroutkova K, Patkova J, Elkalaf M, Dzupa V, Trnka J, Duska F. Normalizing Glutamine Concentration Causes Mitochondrial Uncoupling in an In Vitro Model of Human Skeletal Muscle. JPEN J Parenter Enteral Nutr 2015; 39:180-189. [DOI: 10.1177/0148607113513801] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Adela Krajcova
- Laboratory for Metabolism and Bioenergetics, Department of Nutrition, Third Faculty of Medicine, Charles University in Prague, Czech Republic
| | - Jakub Ziak
- Laboratory for Metabolism and Bioenergetics, Department of Nutrition, Third Faculty of Medicine, Charles University in Prague, Czech Republic
| | - Katerina Jiroutkova
- Laboratory for Metabolism and Bioenergetics, Department of Nutrition, Third Faculty of Medicine, Charles University in Prague, Czech Republic
| | - Jana Patkova
- Laboratory for Metabolism and Bioenergetics, Department of Nutrition, Third Faculty of Medicine, Charles University in Prague, Czech Republic
| | - Moustafa Elkalaf
- Laboratory for Metabolism and Bioenergetics, Department of Nutrition, Third Faculty of Medicine, Charles University in Prague, Czech Republic
| | - Valer Dzupa
- Department of Orthopaedic Surgery, Third Faculty of Medicine, Charles University in Prague, Czech Republic
| | - Jan Trnka
- Laboratory for Metabolism and Bioenergetics, Department of Nutrition, Third Faculty of Medicine, Charles University in Prague, Czech Republic
| | - Frantisek Duska
- Laboratory for Metabolism and Bioenergetics, Department of Nutrition, Third Faculty of Medicine, Charles University in Prague, Czech Republic
| |
Collapse
|
186
|
Takeuchi T, Schumacker PT, Kozmin SA. Identification of fumarate hydratase inhibitors with nutrient-dependent cytotoxicity. J Am Chem Soc 2015; 137:564-7. [PMID: 25469852 PMCID: PMC4308746 DOI: 10.1021/ja5101257] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Indexed: 01/09/2023]
Abstract
Development of cell-permeable small molecules that target enzymes involved in energy metabolism remains important yet challenging. We describe here the discovery of a new class of compounds with a nutrient-dependent cytotoxicity profile that arises from pharmacological inhibition of fumarate hydratase (also known as fumarase). This finding was enabled by a high-throughput screen of a diverse chemical library in a panel of human cancer cell lines cultured under different growth conditions, followed by subsequent structure-activity optimization and target identification. While the highest cytotoxicity was observed under low glucose concentrations, the antiproliferative activities and inhibition of oxygen consumption rates in cells were distinctly different from those displayed by typical inhibitors of mitochondrial oxidative phosphorylation. The use of a photoaffinity labeling strategy identified fumarate hydratase as the principal pharmacological target. Final biochemical studies confirmed dose-dependent, competitive inhibition of this enzyme in vitro, which was fully consistent with the initially observed growth inhibitory activity. Our work demonstrates how the phenotypic observations combined with a successful target identification strategy can yield a useful class of pharmacological inhibitors of an enzyme involved in the operation of tricarboxylic acid cycle.
Collapse
Affiliation(s)
- Toshifumi Takeuchi
- Department
of Chemistry, University of Chicago, Chicago, Illinois 60637, United States
| | - Paul T. Schumacker
- Department
of Pediatrics, Division of Neonatology, Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Sergey A. Kozmin
- Department
of Chemistry, University of Chicago, Chicago, Illinois 60637, United States
| |
Collapse
|
187
|
Abstract
Assessing the bioenergetics of human pluripotent stem cells (hPSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), provides considerable insight into their mitochondrial functions and cellular properties. This might allow exposing potential energetic defects caused by mitochondrial diseases. However, certain challenges have to be met due to unique growth conditions in highly specialized and costly culture media. Here, we describe a method that facilitates the assessment of the bioenergetic profiles of hPSCs in a noninvasive fashion, while requiring only small sample sizes and allowing for several replicates. Basal respiratory and glycolytic capacities are assessed using a XF24 Extracellular Flux Analyzer by simultaneous measurements of the oxygen consumption rate (OCR) and extracellular acidification rate (ECAR), respectively. In addition, bioenergetic parameters are estimated by monitoring OCR and ECAR values upon metabolic perturbations via the consecutive introduction of mitochondria-specific inhibitors.
Collapse
|
188
|
Pham T, Zand K, Wallace D, Burke P. Fluorescence analysis of single mitochondria with nanofluidic channels. Methods Mol Biol 2015; 1264:35-46. [PMID: 25631001 DOI: 10.1007/978-1-4939-2257-4_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Single mitochondrial assays are uncovering a new level of biological heterogeneity, holding promises for a better understanding of molecular respiration and mitochondria-related diseases. Here, we present a nanoscale approach to trapping single mitochondria in fluidic channels for fluorescence microscopy. We fabricate the nanofluidic channels in polydimethylsiloxane and bond them onto a glass slide, creating a highly reproducible device that can be connected to external pumps and mounted to a microscope. Having a unique nanoscale cross section, our channels can trap single mitochondria from a purified mitochondrial preparation flown across. Compared with the traditional fluorescence method to monitor single mitochondrial membrane potential with glass slides and open fluidic chambers, our nanofluidic channels reduce background fluorescence, enhance focus, and allow ease in experimental buffer exchanges. Hence, our channels offer researchers a new effective platform to test their hypotheses on single mitochondria.
Collapse
Affiliation(s)
- Ted Pham
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| | | | | | | |
Collapse
|
189
|
Hasnain SZ, Borg DJ, Harcourt BE, Tong H, Sheng YH, Ng CP, Das I, Wang R, Chen ACH, Loudovaris T, Kay TW, Thomas HE, Whitehead JP, Forbes JM, Prins JB, McGuckin MA. Glycemic control in diabetes is restored by therapeutic manipulation of cytokines that regulate beta cell stress. Nat Med 2014; 20:1417-26. [PMID: 25362253 DOI: 10.1038/nm.3705] [Citation(s) in RCA: 191] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 08/30/2014] [Indexed: 02/07/2023]
Abstract
In type 2 diabetes, hyperglycemia is present when an increased demand for insulin, typically due to insulin resistance, is not met as a result of progressive pancreatic beta cell dysfunction. This defect in beta cell activity is typically characterized by impaired insulin biosynthesis and secretion, usually accompanied by oxidative and endoplasmic reticulum (ER) stress. We demonstrate that multiple inflammatory cytokines elevated in diabetic pancreatic islets induce beta cell oxidative and ER stress, with interleukin-23 (IL-23), IL-24 and IL-33 being the most potent. Conversely, we show that islet-endogenous and exogenous IL-22, by regulating oxidative stress pathways, suppresses oxidative and ER stress caused by cytokines or glucolipotoxicity in mouse and human beta cells. In obese mice, antibody neutralization of IL-23 or IL-24 partially reduced beta cell ER stress and improved glucose tolerance, whereas IL-22 administration modulated oxidative stress regulatory genes in islets, suppressed ER stress and inflammation, promoted secretion of high-quality efficacious insulin and fully restored glucose homeostasis followed by restitution of insulin sensitivity. Thus, therapeutic manipulation of immune regulators of beta cell stress reverses the hyperglycemia central to diabetes pathology.
Collapse
Affiliation(s)
- Sumaira Z Hasnain
- Mucosal Diseases Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Danielle J Borg
- Glycation &Diabetes Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Brooke E Harcourt
- Glycation &Diabetes Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Hui Tong
- Mucosal Diseases Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Yonghua H Sheng
- Mucosal Diseases Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Choa Ping Ng
- Metabolic Medicine Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Indrajit Das
- Mucosal Diseases Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Ran Wang
- Mucosal Diseases Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Alice C-H Chen
- Mucosal Diseases Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | | | - Thomas W Kay
- St. Vincent's Research Institute, Melbourne, Victoria, Australia
| | - Helen E Thomas
- St. Vincent's Research Institute, Melbourne, Victoria, Australia
| | - Jonathan P Whitehead
- 1] Metabolic Medicine Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia. [2] School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Josephine M Forbes
- 1] Glycation &Diabetes Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia. [2] School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Johannes B Prins
- 1] Metabolic Medicine Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia. [2] School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Michael A McGuckin
- 1] Mucosal Diseases Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia. [2] School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia. [3] School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
190
|
Bhattacharya K, Bag AK, Tripathi R, Samanta SK, Pal BC, Shaha C, Mandal C. Mahanine, a novel mitochondrial complex-III inhibitor induces G0/G1 arrest through redox alteration-mediated DNA damage response and regresses glioblastoma multiforme. Am J Cancer Res 2014; 4:629-647. [PMID: 25520856 PMCID: PMC4266700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 09/30/2014] [Indexed: 06/04/2023] Open
Abstract
The Electron transport chain (ETC) is responsible for oxidative phosphorylation-mediated mitochondrial respiration. Here we wanted to address the mahanine-induced targeted pathways in glioblastoma multiforme (GBM) in the context of G0/G1 phase arrest and redox alteration. We have demonstrated mahanine, as a novel mitochondrial complex-III inhibitor which induced G0/G1 phase arrest in GBM. This event was preceded by accumulation of intracellular ROS by the inhibition of mitochondrial ETC. The accumulated ROS induced DNA damage response (DDR), that mediated Chk1/Chk2 upregulation and activation which were essential factors for the G0/G1 arrest. NAC-mediated scavenging of ROS generation reduced the propensity of G0/G1 phase arrest in GBM cells by mahanine. Knockdown of Chk1/Chk2 also affected the cell cycle inhibitory potential of mahanine. During G0/G1 arrest, other hallmark proteins like, cyclin D1/cyclin D3, CDK4/CDK6 and CDC25A were also downregulated. The G0/G1 phase restriction property of mahanine was also established in in vivo mice model. Mahanine-induced complex-III inhibition triggered enhanced ROS in hypoxia responsible for higher G0/G1 arrest. Furthermore, we demonstrated that mahanine-treated G0/G1 arrested cells were less potent to form xenograft tumor in vivo. Additionally, they exhibited reduced ability to migrate and form intracellular tube-like structures. Moreover, they became susceptible to differentiate and astrocyte-like cells were generated from the epithelial lineage. Taken together, our results established that complex-III of ETC is one of the possible potential targets of mahanine. This nontoxic chemotherapeutic molecule enhanced ROS production, induced cell cycle arrest and thereafter regressed GBM without effecting normal astrocytes.
Collapse
Affiliation(s)
- Kaushik Bhattacharya
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology4, Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, India
| | - Arup K Bag
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology4, Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, India
| | - Rakshamani Tripathi
- Cell Death and Differentiation Research, National Institute of ImmunologyNew Delhi, India
| | - Suman K Samanta
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology4, Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, India
| | - Bikas C Pal
- National Institute of Pharmaceutical Education and ResearchKolkata, India
| | - Chandrima Shaha
- Cell Death and Differentiation Research, National Institute of ImmunologyNew Delhi, India
| | - Chitra Mandal
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology4, Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, India
| |
Collapse
|
191
|
Pike Winer LS, Wu M. Rapid analysis of glycolytic and oxidative substrate flux of cancer cells in a microplate. PLoS One 2014; 9:e109916. [PMID: 25360519 PMCID: PMC4215881 DOI: 10.1371/journal.pone.0109916] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Accepted: 09/01/2014] [Indexed: 12/15/2022] Open
Abstract
Cancer cells exhibit remarkable alterations in cellular metabolism, particularly in their nutrient substrate preference. We have devised several experimental methods that rapidly analyze the metabolic substrate flux in cancer cells: glycolysis and the oxidation of major fuel substrates glucose, glutamine, and fatty acids. Using the XF Extracellular Flux analyzer, these methods measure, in real-time, the oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) of living cells in a microplate as they respond to substrates and metabolic perturbation agents. In proof-of-principle experiments, we analyzed substrate flux and mitochondrial bioenergetics of two human glioblastoma cell lines, SF188s and SF188f, which were derived from the same parental cell line but proliferate at slow and fast rates, respectively. These analyses led to three interesting observations: 1) both cell lines respired effectively with substantial endogenous substrate respiration; 2) SF188f cells underwent a significant shift from glycolytic to oxidative metabolism, along with a high rate of glutamine oxidation relative to SF188s cells; and 3) the mitochondrial proton leak-linked respiration of SF188f cells increased significantly compared to SF188s cells. It is plausible that the proton leak of SF188f cells may play a role in allowing continuous glutamine-fueled anaplerotic TCA cycle flux by partially uncoupling the TCA cycle from oxidative phosphorylation. Taken together, these rapid, sensitive and high-throughput substrate flux analysis methods introduce highly valuable approaches for developing a greater understanding of genetic and epigenetic pathways that regulate cellular metabolism, and the development of therapies that target cancer metabolism.
Collapse
Affiliation(s)
- Lisa S Pike Winer
- Seahorse Bioscience Inc., North Billerica, Massachusetts, United States of America
| | - Min Wu
- Seahorse Bioscience Inc., North Billerica, Massachusetts, United States of America
| |
Collapse
|
192
|
Fried NT, Moffat C, Seifert EL, Oshinsky ML. Functional mitochondrial analysis in acute brain sections from adult rats reveals mitochondrial dysfunction in a rat model of migraine. Am J Physiol Cell Physiol 2014; 307:C1017-30. [PMID: 25252946 DOI: 10.1152/ajpcell.00332.2013] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mitochondrial dysfunction has been implicated in many neurological disorders that only develop or are much more severe in adults, yet no methodology exists that allows for medium-throughput functional mitochondrial analysis of brain sections from adult animals. We developed a technique for quantifying mitochondrial respiration in acutely isolated adult rat brain sections with the Seahorse XF Analyzer. Evaluating a range of conditions made quantifying mitochondrial function from acutely derived adult brain sections from the cortex, cerebellum, and trigeminal nucleus caudalis possible. Optimization of this technique demonstrated that the ideal section size was 1 mm wide. We found that sectioning brains at physiological temperatures was necessary for consistent metabolic analysis of trigeminal nucleus caudalis sections. Oxygen consumption in these sections was highly coupled to ATP synthesis, had robust spare respiratory capacities, and had limited nonmitochondrial respiration, all indicative of healthy tissue. We demonstrate the effectiveness of this technique by identifying a decreased spare respiratory capacity in the trigeminal nucleus caudalis of a rat model of chronic migraine, a neurological disorder that has been associated with mitochondrial dysfunction. This technique allows for 24 acutely isolated sections from multiple brain regions of a single adult rat to be analyzed simultaneously with four sequential drug treatments, greatly advancing the ability to study mitochondrial physiology in adult neurological disorders.
Collapse
Affiliation(s)
- Nathan T Fried
- Thomas Jefferson University, Department of Neurology, Philadelphia, Pennsylvania
| | - Cynthia Moffat
- Thomas Jefferson University, Department of Pathology, Anatomy and Cell Biology, Philadelphia, Pennsylvania
| | - Erin L Seifert
- Thomas Jefferson University, Department of Pathology, Anatomy and Cell Biology, Philadelphia, Pennsylvania
| | - Michael L Oshinsky
- Thomas Jefferson University, Department of Neurology, Philadelphia, Pennsylvania;
| |
Collapse
|
193
|
Shen Y, Tian Y, Yang J, Shi X, Ouyang L, Gao J, Lu J. Dual effects of carnosine on energy metabolism of cultured cortical astrocytes under normal and ischemic conditions. ACTA ACUST UNITED AC 2014; 192-193:45-52. [PMID: 25195162 DOI: 10.1016/j.regpep.2014.08.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 07/09/2014] [Accepted: 08/11/2014] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the effects of carnosine on the bioenergetic profile of cultured cortical astrocytes under normal and ischemic conditions. METHODS The Seahorse Bioscience XF96 Extracellular Flux Analyzer was used to measure the oxygen consumption rates (OCRs) and extracellular acidification rates (ECARs) of cultured cortical astrocytes treated with and without carnosine under normal and ischemic conditions. RESULTS Under the normal growth condition, the basal OCRs and ECARs of astrocytes were 21.72±1.59 pmol/min/μg protein and 3.95±0.28 mpH/min/μg protein respectively. Mitochondrial respiration accounted for ~80% of the total cellular respiration and 85% of this coupled to ATP synthesis. Carnosine significantly reduced basal OCRs and ECARs and ATP-linked respiration, but it strikingly increased the spare respiratory capacity of astrocytes. The cellular ATP level in carnosine-treated astrocytes was reduced to ~42% of the control. However, under the ischemic condition, carnosine upregulated the mitochondrial respiratory and cellular ATP content of astrocytes exposed to 8h of oxygen-glucose deprivation (OGD) followed by 24 h of recovery under the normal growth condition. CONCLUSIONS Carnosine may be an endogenous regulator of astrocyte energy metabolism and a clinically safe therapeutic agent for promoting brain energy metabolism recovery after ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Yao Shen
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Yueyang Tian
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jianbo Yang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Xiaojie Shi
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Li Ouyang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jieqiong Gao
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jianxin Lu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
194
|
Zacherl JR, Mihalik SJ, Chace DH, Christensen TC, Robinson LJ, Blair HC. Elaidate, an 18-carbon trans-monoenoic fatty acid, inhibits β-oxidation in human peripheral blood macrophages. J Cell Biochem 2014; 115:62-70. [PMID: 23904193 DOI: 10.1002/jcb.24633] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/22/2013] [Indexed: 12/11/2022]
Abstract
Consumption of trans-unsaturated fatty acids promotes atherosclerosis, but whether degradation of fats in macrophages is altered by trans-unsaturated fatty acids is unknown. We compared the metabolism of oleate (C18:1Δ9-10 cis; (Z)-octadec-9-enoate), elaidate (C18:Δ9-10 trans; (E)-octadec-9-enoate), and stearate (C18:0, octadecanoate) in adherent peripheral human macrophages. Metabolism was followed by measurement of acylcarnitines in cell supernatants by MS/MS, determination of cellular fatty acid content by GC/MS, and assessment of β-oxidation rates using radiolabeled fatty acids. Cells incubated for 44 h in 100 µM elaidate accumulated more unsaturated fatty acids, including both longer- and shorter-chain, and had reduced C18:0 relative to those incubated with oleate or stearate. Both C12:1 and C18:1 acylcarnitines accumulated in supernatants of macrophages exposed to trans fats. These results suggested β-oxidation inhibition one reaction proximal to the trans bond. Comparison of [1-(14)C]oleate to [1-(14)C]elaidate catabolism showed that elaidate completed the first round of fatty acid β-oxidation at rates comparable to oleate. Yet, in competitive β-oxidation assays with [9,10-(3)H]oleate, tritium release rate decreased when unlabeled oleate was replaced by the same quantity of elaidate. These data show specific inhibition of monoenoic fat catabolism by elaidate that is not shared by other atherogenic fats.
Collapse
Affiliation(s)
- Janelle R Zacherl
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261
| | | | | | | | | | | |
Collapse
|
195
|
Barbi de Moura M, Uppala R, Zhang Y, Van Houten B, Goetzman ES. Overexpression of mitochondrial sirtuins alters glycolysis and mitochondrial function in HEK293 cells. PLoS One 2014; 9:e106028. [PMID: 25165814 PMCID: PMC4148395 DOI: 10.1371/journal.pone.0106028] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 07/30/2014] [Indexed: 01/15/2023] Open
Abstract
SIRT3, SIRT4, and SIRT5 are mitochondrial deacylases that impact multiple facets of energy metabolism and mitochondrial function. SIRT3 activates several mitochondrial enzymes, SIRT4 represses its targets, and SIRT5 has been shown to both activate and repress mitochondrial enzymes. To gain insight into the relative effects of the mitochondrial sirtuins in governing mitochondrial energy metabolism, SIRT3, SIRT4, and SIRT5 overexpressing HEK293 cells were directly compared. When grown under standard cell culture conditions (25 mM glucose) all three sirtuins induced increases in mitochondrial respiration, glycolysis, and glucose oxidation, but with no change in growth rate or in steady-state ATP concentration. Increased proton leak, as evidenced by oxygen consumption in the presence of oligomycin, appeared to explain much of the increase in basal oxygen utilization. Growth in 5 mM glucose normalized the elevations in basal oxygen consumption, proton leak, and glycolysis in all sirtuin over-expressing cells. While the above effects were common to all three mitochondrial sirtuins, some differences between the SIRT3, SIRT4, and SIRT5 expressing cells were noted. Only SIRT3 overexpression affected fatty acid metabolism, and only SIRT4 overexpression altered superoxide levels and mitochondrial membrane potential. We conclude that all three mitochondrial sirtuins can promote increased mitochondrial respiration and cellular metabolism. SIRT3, SIRT4, and SIRT5 appear to respond to excess glucose by inducing a coordinated increase of glycolysis and respiration, with the excess energy dissipated via proton leak.
Collapse
Affiliation(s)
- Michelle Barbi de Moura
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Radha Uppala
- Division of Medical Genetics, Department of Pediatrics, Children’s Hospital of Pittsburgh of The University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Yuxun Zhang
- Division of Medical Genetics, Department of Pediatrics, Children’s Hospital of Pittsburgh of The University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Bennett Van Houten
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, United States of America
| | - Eric S. Goetzman
- Division of Medical Genetics, Department of Pediatrics, Children’s Hospital of Pittsburgh of The University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
196
|
Shen Y, Yang J, Li J, Shi X, Ouyang L, Tian Y, Lu J. Carnosine inhibits the proliferation of human gastric cancer SGC-7901 cells through both of the mitochondrial respiration and glycolysis pathways. PLoS One 2014; 9:e104632. [PMID: 25115854 PMCID: PMC4130552 DOI: 10.1371/journal.pone.0104632] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 07/15/2014] [Indexed: 12/19/2022] Open
Abstract
Carnosine, a naturally occurring dipeptide, has been recently demonstrated to possess anti-tumor activity. However, its underlying mechanism is unclear. In this study, we investigated the effect and mechanism of carnosine on the cell viability and proliferation of the cultured human gastric cancer SGC-7901 cells. Carnosine treatment did not induce cell apoptosis or necrosis, but reduced the proliferative capacity of SGC-7901 cells. Seahorse analysis showed SGC-7901 cells cultured with pyruvate have active mitochondria, and depend on mitochondrial oxidative phosphorylation more than glycolysis pathway for generation of ATP. Carnosine markedly decreased the absolute value of mitochondrial ATP-linked respiration, and reduced the maximal oxygen consumption and spare respiratory capacity, which may reduce mitochondrial function correlated with proliferative potential. Simultaneously, carnosine also reduced the extracellular acidification rate and glycolysis of SGC-7901 cells. Our results suggested that carnosine is a potential regulator of energy metabolism of SGC-7901 cells both in the anaerobic and aerobic pathways, and provided a clue for preclinical and clinical evaluation of carnosine for gastric cancer therapy.
Collapse
Affiliation(s)
- Yao Shen
- Key Laboratory of Laboratory Medicine, Ministry of Education, Attardi Institute of Mitochondrial Biomedicine and Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, China
- * E-mail: (YS); (JXL)
| | - Jianbo Yang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Attardi Institute of Mitochondrial Biomedicine and Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, China
| | - Juan Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, Attardi Institute of Mitochondrial Biomedicine and Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaojie Shi
- Key Laboratory of Laboratory Medicine, Ministry of Education, Attardi Institute of Mitochondrial Biomedicine and Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, China
| | - Li Ouyang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Attardi Institute of Mitochondrial Biomedicine and Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, China
| | - Yueyang Tian
- Key Laboratory of Laboratory Medicine, Ministry of Education, Attardi Institute of Mitochondrial Biomedicine and Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, China
| | - Jianxin Lu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Attardi Institute of Mitochondrial Biomedicine and Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, China
- * E-mail: (YS); (JXL)
| |
Collapse
|
197
|
Tumorigenicity of hypoxic respiring cancer cells revealed by a hypoxia-cell cycle dual reporter. Proc Natl Acad Sci U S A 2014; 111:12486-91. [PMID: 25114222 DOI: 10.1073/pnas.1402012111] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Although aerobic glycolysis provides an advantage in the hypoxic tumor microenvironment, some cancer cells can also respire via oxidative phosphorylation. These respiring ("non-Warburg") cells were previously thought not to play a key role in tumorigenesis and thus fell from favor in the literature. We sought to determine whether subpopulations of hypoxic cancer cells have different metabolic phenotypes and gene-expression profiles that could influence tumorigenicity and therapeutic response, and we therefore developed a dual fluorescent protein reporter, HypoxCR, that detects hypoxic [hypoxia-inducible factor (HIF) active] and/or cycling cells. Using HEK293T cells as a model, we identified four distinct hypoxic cell populations by flow cytometry. The non-HIF/noncycling cell population expressed a unique set of genes involved in mitochondrial function. Relative to the other subpopulations, these hypoxic "non-Warburg" cells had highest oxygen consumption rates and mitochondrial capacity consistent with increased mitochondrial respiration. We found that these respiring cells were unexpectedly tumorigenic, suggesting that continued respiration under limiting oxygen conditions may be required for tumorigenicity.
Collapse
|
198
|
Shen Y, Tian Y, Shi X, Yang J, Ouyang L, Gao J, Lu J. Exposure to high glutamate concentration activates aerobic glycolysis but inhibits ATP-linked respiration in cultured cortical astrocytes. Cell Biochem Funct 2014; 32:530-7. [PMID: 25077445 DOI: 10.1002/cbf.3047] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 06/18/2014] [Accepted: 06/23/2014] [Indexed: 11/06/2022]
Abstract
Astrocytes play a key role in removing the synaptically released glutamate from the extracellular space and maintaining the glutamate below neurotoxic level in the brain. However, high concentration of glutamate leads to toxicity in astrocytes, and the underlying mechanisms are unclear. The purpose of this study was to investigate whether energy metabolism disorder, especially impairment of mitochondrial respiration, is involved in the glutamate-induced gliotoxicity. Exposure to 10-mM glutamate for 48 h stimulated glycolysis and respiration in astrocytes. However, the increased oxygen consumption was used for proton leak and non-mitochondrial respiration, but not for oxidative phosphorylation and ATP generation. When the exposure time extended to 72 h, glycolysis was still activated for ATP generation, but the mitochondrial ATP-linked respiration of astrocytes was reduced. The glutamate-induced astrocyte damage can be mimicked by the non-metabolized substrate d-aspartate but reversed by the non-selective glutamate transporter inhibitor TBOA. In addition, the glutamate toxicity can be partially reversed by vitamin E. These findings demonstrate that changes of bioenergetic profile occur in cultured cortical astrocytes exposed to high concentration of glutamate and highlight the role of mitochondria respiration in glutamate-induced gliotoxicity in cortical astrocytes.
Collapse
Affiliation(s)
- Yao Shen
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | | | | | | | | | | | | |
Collapse
|
199
|
Kitani T, Kami D, Matoba S, Gojo S. Internalization of isolated functional mitochondria: involvement of macropinocytosis. J Cell Mol Med 2014; 18:1694-703. [PMID: 24912369 PMCID: PMC4190914 DOI: 10.1111/jcmm.12316] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 04/03/2014] [Indexed: 12/03/2022] Open
Abstract
In eukaryotic cells, mitochondrial dysfunction is associated with a variety of human diseases. Delivery of exogenous functional mitochondria into damaged cells has been proposed as a mechanism of cell transplant and physiological repair for damaged tissue. We here demonstrated that isolated mitochondria can be transferred into homogeneic and xenogeneic cells by simple co-incubation using genetically labelled mitochondria, and elucidated the mechanism and the effect of direct mitochondrial transfer. Intracellular localization of exogenous mitochondria was confirmed by PCR, real-time PCR, live fluorescence imaging, three-dimensional reconstruction imaging, continuous time-lapse microscopic observation, flow cytometric analysis and immunoelectron microscopy. Isolated homogeneic mitochondria were transferred into human uterine endometrial gland-derived mesenchymal cells in a dose-dependent manner. Moreover, mitochondrial transfer rescued the mitochondrial respiratory function and improved the cellular viability in mitochondrial DNA-depleted cells and these effects lasted several days. Finally, we discovered that mitochondrial internalization involves macropinocytosis. In conclusion, these data support direct transfer of exogenous mitochondria as a promising approach for the treatment of various diseases.
Collapse
Affiliation(s)
- Tomoya Kitani
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University School of Medicine, Kyoto, Japan
| | | | | | | |
Collapse
|
200
|
Le TT, Urasaki Y, Pizzorno G. Uridine prevents tamoxifen-induced liver lipid droplet accumulation. BMC Pharmacol Toxicol 2014; 15:27. [PMID: 24887406 PMCID: PMC4064512 DOI: 10.1186/2050-6511-15-27] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 04/30/2014] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Tamoxifen, an agonist of estrogen receptor, is widely prescribed for the prevention and long-term treatment of breast cancer. A side effect of tamoxifen is fatty liver, which increases the risk for non-alcoholic fatty liver disease. Prevention of tamoxifen-induced fatty liver has the potential to improve the safety of long-term tamoxifen usage. METHODS Uridine, a pyrimidine nucleoside with reported protective effects against drug-induced fatty liver, was co-administered with tamoxifen in C57BL/6J mice. Liver lipid levels were evaluated with lipid visualization using coherent anti-Stokes Raman scatting (CARS) microscopy, biochemical assay measurement of triacylglyceride (TAG), and liquid chromatography coupled with mass spectrometry (LC-MS) measurement of membrane phospholipid. Blood TAG and cholesterol levels were measured. Mitochondrial respiration of primary hepatocytes in the presence of tamoxifen and/or uridine was evaluated by measuring oxygen consumption rate with an extracellular flux analyzer. Liver protein lysine acetylation profiles were evaluated with 1D and 2D Western blots. In addition, the relationship between endogenous uridine levels, fatty liver, and tamoxifen administration was evaluated in transgenic mice UPase1-/-and UPase1-TG. RESULTS Uridine co-administration prevented tamoxifen-induced liver lipid droplet accumulation in mice. The most prominent effect of uridine co-administration with tamoxifen was the stimulation of liver membrane phospholipid biosynthesis. Uridine had no protective effect against tamoxifen-induced impairment to mitochondrial respiration of primary hepatocytes or liver TAG and cholesterol export. Uridine had no effect on tamoxifen-induced changes to liver protein acetylation profile. Transgenic mice UPase1-/-with increased pyrimidine salvage activity were protected against tamoxifen-induced liver lipid droplet accumulation. In contrast, UPase1-TG mice with increased pyrimidine catabolism activity had intrinsic liver lipid droplet accumulation, which was aggravated following tamoxifen administration. CONCLUSION Uridine co-administration was effective at preventing tamoxifen-induced liver lipid droplet accumulation. The ability of uridine to prevent tamoxifen-induced fatty liver appeared to depend on the pyrimidine salvage pathway, which promotes biosynthesis of membrane phospholipid.
Collapse
Affiliation(s)
- Thuc T Le
- Nevada Cancer Institute, One Breakthrough Way, Las Vegas, NV 89135, USA.
| | | | | |
Collapse
|