1
|
Kim S, Park JG, Choi SH, Kim JW, Jin MS. Cryo-EM structures reveal the H +/citrate symport mechanism of Drosophila INDY. Life Sci Alliance 2025; 8:e202402992. [PMID: 39884835 PMCID: PMC11782487 DOI: 10.26508/lsa.202402992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 02/01/2025] Open
Abstract
Drosophila I'm Not Dead Yet (INDY) functions as a transporter for citrate, a key metabolite in the citric acid cycle, across the plasma membrane. Partial deficiency of INDY extends lifespan, akin to the effects of caloric restriction. In this work, we use cryo-electron microscopy to determine structures of INDY in the presence and absence of citrate and in complex with the well-known inhibitor 4,4'-diisothiocyano-2,2'-disulfonic acid stilbene (DIDS) at resolutions ranging from 2.7 to 3.6 Å. Together with functional data obtained in vitro, the INDY structures reveal the H+/citrate co-transport mechanism, in which aromatic residue F119 serves as a one-gate element. They also provide insight into how protein-lipid interactions at the dimerization interface affect the stability and function of the transporter, and how DIDS disrupts the transport cycle.
Collapse
Affiliation(s)
- Subin Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Jun Gyou Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Seung Hun Choi
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Ji Won Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Mi Sun Jin
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| |
Collapse
|
2
|
Jaramillo-Martinez V, Sennoune SR, Tikhonova EB, Karamyshev AL, Ganapathy V, Urbatsch IL. Molecular phenotypes segregate missense mutations in SLC13A5 Epilepsy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.23.594637. [PMID: 38826402 PMCID: PMC11142175 DOI: 10.1101/2024.05.23.594637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The sodium-coupled citrate transporter (NaCT, SLC13A5) mediates citrate uptake across the plasma membrane via an inward Na + gradient. Mutations in SLC13A5 cause early infantile epileptic encephalopathy type-25 (EIEE25, SLC13A5 Epilepsy) due to impaired citrate uptake in neurons. Despite clinical identification of disease-causing mutations, underlying mechanisms and cures remain elusive. We mechanistically classify the molecular phenotypes of six mutations. C50R, T142M, and T227M exhibit impaired citrate transport despite normal expression at the cell surface. G219R, S427L, and L488P are hampered by low protein expression, ER retention, and reduced transport. Mutants' mRNA levels resemble wildtype, suggesting post-translational defects. Class II mutations display immature core-glycosylation and shortened half-lives, indicating protein folding defects. These experiments provide a comprehensive understanding of the mutation's defects in SLC13A5 Epilepsy at the biochemical and molecular level and shed light into the trafficking pathway(s) of NaCT. The two classes of mutations will require fundamentally different treatment approaches to either restore transport function, or enable correction of protein folding defects. Summary Loss-of-function mutations in the SLC13A5 causes SLC13A5-Epilepsy, a devastating disease characterized by neonatal epilepsy. Currently no cure is available. We clarify the molecular-level defects to guide future developments for phenotype-specific treatment of disease-causing mutations.
Collapse
|
3
|
Mishra D, Kannan K, Meadows K, Macro J, Li M, Frankel S, Rogina B. INDY-From Flies to Worms, Mice, Rats, Non-Human Primates, and Humans. FRONTIERS IN AGING 2022; 2:782162. [PMID: 35822025 PMCID: PMC9261455 DOI: 10.3389/fragi.2021.782162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/24/2021] [Indexed: 01/17/2023]
Abstract
I’m Not Dead Yet (Indy) is a fly homologue of the mammalian SLC13A5 (mSLC13A5) plasma membrane citrate transporter, a key metabolic regulator and energy sensor involved in health, longevity, and disease. Reduction of Indy gene activity in flies, and its homologs in worms, modulates metabolism and extends longevity. The metabolic changes are similar to what is obtained with caloric restriction (dietary restriction). Similar effects on metabolism have been observed in mice and rats. As a citrate transporter, INDY regulates cytoplasmic citrate levels. Indy flies heterozygous for a P-element insertion have increased spontaneous physical activity, increased fecundity, reduced insulin signaling, increased mitochondrial biogenesis, preserved intestinal stem cell homeostasis, lower lipid levels, and increased stress resistance. Mammalian Indy knockout (mIndy-KO) mice have higher sensitivity to insulin signaling, lower blood pressure and heart rate, preserved memory and are protected from the negative effects of a high-fat diet and some of the negative effects of aging. Reducing mIndy expression in human hepatocarcinoma cells has recently been shown to inhibit cell proliferation. Reduced Indy expression in the fly intestine affects intestinal stem cell proliferation, and has recently been shown to also inhibit germ cell proliferation in males with delayed sperm maturation and decreased spermatocyte numbers. These results highlight a new connection between energy metabolism and cell proliferation. The overrall picture in a variety of species points to a conserved role of INDY for metabolism and health. This is illustrated by an association of high mIndy gene expression with non-alcoholic fatty liver disease in obese humans. mIndy (mSLC13A5) coding region mutations (e.g., loss-of-function) are also associated with adverse effects in humans, such as autosomal recessive early infantile epileptic encephalopathy and Kohlschütter−Tönz syndrome. The recent findings illustrate the importance of mIndy gene for human health and disease. Furthermore, recent work on small-molecule regulators of INDY highlights the promise of INDY-based treatments for ameliorating disease and promoting healthy aging.
Collapse
Affiliation(s)
- Dushyant Mishra
- Department of Genetics and Genome Sciences, School of Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Kavitha Kannan
- Department of Genetics and Genome Sciences, School of Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Kali Meadows
- Department of Genetics and Genome Sciences, School of Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Jacob Macro
- Department of Genetics and Genome Sciences, School of Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Michael Li
- Department of Genetics and Genome Sciences, School of Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Stewart Frankel
- Department of Biology, University of Hartford, West Hartford, CT, United States
| | - Blanka Rogina
- Department of Genetics and Genome Sciences, School of Medicine, University of Connecticut Health Center, Farmington, CT, United States.,Institute for Systems Genomics, School of Medicine, University of Connecticut Health Center, Farmington, CT, United States
| |
Collapse
|
4
|
Kannan K, Rogina B. The Role of Citrate Transporter INDY in Metabolism and Stem Cell Homeostasis. Metabolites 2021; 11:705. [PMID: 34677421 PMCID: PMC8540898 DOI: 10.3390/metabo11100705] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
I'm Not Dead Yet (Indy) is a fly gene that encodes a homologue of mammalian SLC13A5 plasma membrane citrate transporter. Reducing expression of Indy gene in flies, and its homologues in worms, extends longevity. Indy reduction in flies, worms, mice and rats affects metabolism by regulating the levels of cytoplasmic citrate, inducing a state similar to calorie restriction. Changes include lower lipid levels, increased insulin sensitivity, increased mitochondrial biogenesis, and prevention of weight gain, among others. The INDY protein is predominantly expressed in fly metabolic tissues: the midgut, fat body and oenocytes. Changes in fly midgut metabolism associated with reduced Indy gene activity lead to preserved mitochondrial function and reduced production of reactive oxygen species. All these changes lead to preserved intestinal stem cell homeostasis, which has a key role in maintaining intestinal epithelium function and enhancing fly healthspan and lifespan. Indy gene expression levels change in response to caloric content of the diet, inflammation and aging, suggesting that INDY regulates metabolic adaptation to nutrition or energetic requirements by controlling citrate levels.
Collapse
Affiliation(s)
- Kavitha Kannan
- Department of Genetics & Genome Sciences, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA;
| | - Blanka Rogina
- Department of Genetics & Genome Sciences, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA;
- Institute for Systems Genomics, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
| |
Collapse
|
5
|
Jaramillo-Martinez V, Sivaprakasam S, Ganapathy V, Urbatsch IL. Drosophila INDY and Mammalian INDY: Major Differences in Transport Mechanism and Structural Features despite Mostly Similar Biological Functions. Metabolites 2021; 11:metabo11100669. [PMID: 34677384 PMCID: PMC8537002 DOI: 10.3390/metabo11100669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
INDY (I’m Not Dead Yet) is a plasma membrane transporter for citrate, first identified in Drosophila. Partial deficiency of INDY extends lifespan in this organism in a manner similar to that of caloric restriction. The mammalian counterpart (NaCT/SLC13A5) also transports citrate. In mice, it is the total, not partial, absence of the transporter that leads to a metabolic phenotype similar to that caloric restriction; however, there is evidence for subtle neurological dysfunction. Loss-of-function mutations in SLC13A5 (solute carrier gene family 13, member A5) occur in humans, causing a recessive disease, with severe clinical symptoms manifested by neonatal seizures and marked disruption in neurological development. Though both Drosophila INDY and mammalian INDY transport citrate, the translocation mechanism differs, the former being a dicarboxylate exchanger for the influx of citrate2− in exchange for other dicarboxylates, and the latter being a Na+-coupled uniporter for citrate2−. Their structures also differ as evident from only ~35% identity in amino acid sequence and from theoretically modeled 3D structures. The varied biological consequences of INDY deficiency across species, with the beneficial effects predominating in lower organisms and detrimental effects overwhelming in higher organisms, are probably reflective of species-specific differences in tissue expression and also in relative contribution of extracellular citrate to metabolic pathways in different tissues
Collapse
Affiliation(s)
- Valeria Jaramillo-Martinez
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - Sathish Sivaprakasam
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (S.S.); (V.G.)
| | - Vadivel Ganapathy
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (S.S.); (V.G.)
| | - Ina L. Urbatsch
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; (S.S.); (V.G.)
- Correspondence:
| |
Collapse
|
6
|
Consequences of NaCT/SLC13A5/mINDY deficiency: good versus evil, separated only by the blood-brain barrier. Biochem J 2021; 478:463-486. [PMID: 33544126 PMCID: PMC7868109 DOI: 10.1042/bcj20200877] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 02/08/2023]
Abstract
NaCT/SLC13A5 is a Na+-coupled transporter for citrate in hepatocytes, neurons, and testes. It is also called mINDY (mammalian ortholog of ‘I'm Not Dead Yet’ in Drosophila). Deletion of Slc13a5 in mice leads to an advantageous phenotype, protecting against diet-induced obesity, and diabetes. In contrast, loss-of-function mutations in SLC13A5 in humans cause a severe disease, EIEE25/DEE25 (early infantile epileptic encephalopathy-25/developmental epileptic encephalopathy-25). The difference between mice and humans in the consequences of the transporter deficiency is intriguing but probably explainable by the species-specific differences in the functional features of the transporter. Mouse Slc13a5 is a low-capacity transporter, whereas human SLC13A5 is a high-capacity transporter, thus leading to quantitative differences in citrate entry into cells via the transporter. These findings raise doubts as to the utility of mouse models to evaluate NaCT biology in humans. NaCT-mediated citrate entry in the liver impacts fatty acid and cholesterol synthesis, fatty acid oxidation, glycolysis, and gluconeogenesis; in neurons, this process is essential for the synthesis of the neurotransmitters glutamate, GABA, and acetylcholine. Thus, SLC13A5 deficiency protects against obesity and diabetes based on what the transporter does in hepatocytes, but leads to severe brain deficits based on what the transporter does in neurons. These beneficial versus detrimental effects of SLC13A5 deficiency are separable only by the blood-brain barrier. Can we harness the beneficial effects of SLC13A5 deficiency without the detrimental effects? In theory, this should be feasible with selective inhibitors of NaCT, which work only in the liver and do not get across the blood-brain barrier.
Collapse
|
7
|
A home run for human NaCT/SLC13A5/INDY: cryo-EM structure and homology model to predict transport mechanisms, inhibitor interactions and mutational defects. Biochem J 2021; 478:2051-2057. [PMID: 34101804 PMCID: PMC8203205 DOI: 10.1042/bcj20210211] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/14/2021] [Accepted: 05/18/2021] [Indexed: 11/26/2022]
Abstract
NaCT (SLC13A5) is a Na+-coupled transporter for citrate, which is expressed in the liver, brain, testes, and bone. It is the mammalian homolog of Drosophila INDY, a cation-independent transporter for citrate, whose partial loss extends lifespan in the organism. In humans, loss-of-function mutations in NaCT cause a disease with severe neurological dysfunction, characterized by neonatal epilepsy and delayed brain development. In contrast with humans, deletion of NaCT in mice results in a beneficial metabolic phenotype with protection against diet-induced obesity and metabolic syndrome; the brain dysfunction is not readily noticeable. The disease-causing mutations are located in different regions of human NaCT protein, suggesting that different mutations might have different mechanisms for the loss of function. The beneficial effects of NaCT loss in the liver versus the detrimental effects of NaCT loss in the brain provide an opportunity to design high-affinity inhibitors for the transporter that do not cross the blood-brain barrier so that only the beneficial effects could be harnessed. To realize these goals, we need a detailed knowledge of the 3D structure of human NaCT. The recent report by Sauer et al. in Nature describing the cryo-EM structure of human NaCT represents such a milestone, paving the way for a better understanding of the structure-function relationship for this interesting and clinically important transporter.
Collapse
|
8
|
Willmes DM, Daniels M, Kurzbach A, Lieske S, Bechmann N, Schumann T, Henke C, El-Agroudy NN, Da Costa Goncalves AC, Peitzsch M, Hofmann A, Kanczkowski W, Kräker K, Müller DN, Morawietz H, Deussen A, Wagner M, El-Armouche A, Helfand SL, Bornstein SR, de Cabo R, Bernier M, Eisenhofer G, Tank J, Jordan J, Birkenfeld AL. The longevity gene mIndy (I'm Not Dead, Yet) affects blood pressure through sympathoadrenal mechanisms. JCI Insight 2021; 6:136083. [PMID: 33491666 PMCID: PMC7934862 DOI: 10.1172/jci.insight.136083] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 12/02/2020] [Indexed: 12/14/2022] Open
Abstract
Reduced expression of the plasma membrane citrate transporter INDY (acronym I’m Not Dead, Yet) extends life span in lower organisms. Deletion of the mammalian Indy (mIndy) gene in rodents improves metabolism via mechanisms akin to caloric restriction, known to lower blood pressure (BP) by sympathoadrenal inhibition. We hypothesized that mIndy deletion attenuates sympathoadrenal support of BP. Continuous arterial BP and heart rate (HR) were reduced in mINDY-KO mice. Concomitantly, urinary catecholamine content was lower, and the decreases in BP and HR by mIndy deletion were attenuated after autonomic ganglionic blockade. Catecholamine biosynthesis pathways were reduced in mINDY-KO adrenals using unbiased microarray analysis. Citrate, the main mINDY substrate, increased catecholamine content in pheochromocytoma cells, while pharmacological inhibition of citrate uptake blunted the effect. Our data suggest that deletion of mIndy reduces sympathoadrenal support of BP and HR by attenuating catecholamine biosynthesis. Deletion of mIndy recapitulates beneficial cardiovascular and metabolic responses to caloric restriction, making it an attractive therapeutic target. Deletion of mIndy reduces blood pressure and heart rate by attenuating catecholamine biosynthesis and recapitulates beneficial cardiovascular and metabolic responses to caloric restriction.
Collapse
Affiliation(s)
- Diana M Willmes
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technical University Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Martin Daniels
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technical University Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, University of Tübingen, Tübingen, Germany.,Department of Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - Anica Kurzbach
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technical University Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, University of Tübingen, Tübingen, Germany.,Department of Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany.,Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Stefanie Lieske
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and
| | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital and Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Tina Schumann
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technical University Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Christine Henke
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technical University Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Nermeen N El-Agroudy
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technical University Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | | | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital and Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Anja Hofmann
- Division of Vascular Endothelium and Microcirculation, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Waldemar Kanczkowski
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Kristin Kräker
- Experimental and Clinical Research Center, Max Delbruck Center for Molecular Medicine and Charité - University Hospital Berlin, Berlin, Germany
| | - Dominik N Müller
- Experimental and Clinical Research Center, Max Delbruck Center for Molecular Medicine and Charité - University Hospital Berlin, Berlin, Germany
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Andreas Deussen
- Department of Physiology, Medical Faculty Carl Gustav Carus, and
| | - Michael Wagner
- Department of Pharmacology and Toxicology, University Hospital and Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Ali El-Armouche
- Department of Pharmacology and Toxicology, University Hospital and Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Stephen L Helfand
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, Rhode Island, USA
| | - Stephan R Bornstein
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Graeme Eisenhofer
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Institute of Clinical Chemistry and Laboratory Medicine, University Hospital and Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Jens Tank
- Aerospace Medicine, University of Cologne, Cologne, Germany
| | - Jens Jordan
- Aerospace Medicine, University of Cologne, Cologne, Germany.,Institute for Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Andreas L Birkenfeld
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technical University Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, University of Tübingen, Tübingen, Germany.,Department of Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany.,Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| |
Collapse
|
9
|
Webb M, Sideris DP. Intimate Relations-Mitochondria and Ageing. Int J Mol Sci 2020; 21:ijms21207580. [PMID: 33066461 PMCID: PMC7589147 DOI: 10.3390/ijms21207580] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial dysfunction is associated with ageing, but the detailed causal relationship between the two is still unclear. We review the major phenomenological manifestations of mitochondrial age-related dysfunction including biochemical, regulatory and energetic features. We conclude that the complexity of these processes and their inter-relationships are still not fully understood and at this point it seems unlikely that a single linear cause and effect relationship between any specific aspect of mitochondrial biology and ageing can be established in either direction.
Collapse
Affiliation(s)
- Michael Webb
- Mitobridge Inc., an Astellas Company, 1030 Massachusetts Ave, Cambridge, MA 02138, USA
| | - Dionisia P Sideris
- Mitobridge Inc., an Astellas Company, 1030 Massachusetts Ave, Cambridge, MA 02138, USA
| |
Collapse
|
10
|
Zhang T, Liu H, Wang J, Li L, Han C, Mustafa A, Xiong X. Evidences in duck (Anas platyrhynchos) by transcriptome data for supporting the biliverdin was mainly synthesized by shell gland. Poult Sci 2019; 98:2260-2271. [DOI: 10.3382/ps/pey576] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 12/19/2018] [Indexed: 12/22/2022] Open
|
11
|
Garcia JF, Carbone MA, Mackay TFC, Anholt RRH. Regulation of Drosophila Lifespan by bellwether Promoter Alleles. Sci Rep 2017; 7:4109. [PMID: 28646164 PMCID: PMC5482829 DOI: 10.1038/s41598-017-04530-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 05/16/2017] [Indexed: 01/24/2023] Open
Abstract
Longevity varies among individuals, but how natural genetic variation contributes to variation in lifespan is poorly understood. Drosophila melanogaster presents an advantageous model system to explore the genetic underpinnings of longevity, since its generation time is brief and both the genetic background and rearing environment can be precisely controlled. The bellwether (blw) gene encodes the α subunit of mitochondrial ATP synthase. Since metabolic rate may influence lifespan, we investigated whether alternative haplotypes in the blw promoter affect lifespan when expressed in a co-isogenic background. We amplified 521 bp upstream promoter sequences containing alternative haplotypes and assessed promoter activity both in vitro and in vivo using a luciferase reporter system. The AG haplotype showed significantly greater expression of luciferase than the GT haplotype. We then overexpressed a blw cDNA construct driven by either the AG or GT haplotype promoter in transgenic flies and showed that the AG haplotype also results in greater blw cDNA expression and a significant decrease in lifespan relative to the GT promoter haplotype, in male flies only. Thus, our results show that naturally occurring regulatory variants of blw affect lifespan in a sex-specific manner.
Collapse
Affiliation(s)
- Júlia Frankenberg Garcia
- Program in Genetics, W. M. Keck Center for Behavioral Biology, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
- School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Mary Anna Carbone
- Program in Genetics, W. M. Keck Center for Behavioral Biology, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Trudy F C Mackay
- Program in Genetics, W. M. Keck Center for Behavioral Biology, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Robert R H Anholt
- Program in Genetics, W. M. Keck Center for Behavioral Biology, Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA.
| |
Collapse
|
12
|
Rogina B. INDY-A New Link to Metabolic Regulation in Animals and Humans. Front Genet 2017; 8:66. [PMID: 28596784 PMCID: PMC5442177 DOI: 10.3389/fgene.2017.00066] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 05/09/2017] [Indexed: 12/02/2022] Open
Abstract
The Indy (I’m Not Dead Yet) gene encodes the fly homolog of the mammalian SLC13A5 citrate transporter. Reduced expression of the Indy gene in flies and worms extends their longevity. INDY is expressed in the plasma membrane of metabolically active tissues. Decreased expression of Indy in worms, flies, mice, and rats alters metabolism in a manner similar to calorie restriction. Reducing INDY activity prevents weight gain in flies, worms, and mice, and counteracts the negative effects of age or a high fat diet on metabolism and insulin sensitivity. The metabolic effects of reducing INDY activity are the result of reduced cytoplasmic citrate. Citrate is a key metabolite and has a central role in energy status of the cell by effecting lipid and carbohydrate metabolism and energy production. Thereby newly described drugs that reduce INDY transporting activity increase insulin sensitivity and reduce hepatic lipid levels via its effect on hepatic citrate uptake. A recent report presented the first direct link between increased hepatic levels of human INDY, insulin resistance, and non-alcoholic fatty liver disease in obese humans. Similarly increased hepatic mIndy levels were observed in non-human primates fed on a high fat diet for 2 years. This effect is mediated via the stimulatory effect of the interleukin-6/Stat3 pathway on mINDY hepatic expression. These findings make INDY a potential and very promising target for the treatment of metabolic disorders in humans.
Collapse
Affiliation(s)
- Blanka Rogina
- Department of Genetics and Genome Sciences, Institute for Systems Genomics, School of Medicine, University of Connecticut Health Center, FarmingtonCT, United States
| |
Collapse
|
13
|
Pesta DH, Perry RJ, Guebre-Egziabher F, Zhang D, Jurczak M, Fischer-Rosinsky A, Daniels MA, Willmes DM, Bhanot S, Bornstein SR, Knauf F, Samuel VT, Shulman GI, Birkenfeld AL. Prevention of diet-induced hepatic steatosis and hepatic insulin resistance by second generation antisense oligonucleotides targeted to the longevity gene mIndy (Slc13a5). Aging (Albany NY) 2016; 7:1086-93. [PMID: 26647160 PMCID: PMC4712334 DOI: 10.18632/aging.100854] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Reducing the expression of the Indy (I'm Not Dead Yet) gene in lower organisms extends life span by mechanisms resembling caloric restriction. Similarly, deletion of the mammalian homolog, mIndy (Slc13a5), encoding for a plasma membrane tricarboxylate transporter, protects from aging- and diet-induced adiposity and insulin resistance in mice. The organ specific contribution to this phenotype is unknown. We examined the impact of selective inducible hepatic knockdown of mIndy on whole body lipid and glucose metabolism using 2′-O-methoxyethyl chimeric anti-sense oligonucleotides (ASOs) in high-fat fed rats. 4-week treatment with 2′-O-methoxyethyl chimeric ASO reduced mIndy mRNA expression by 91% (P<0.001) compared to control ASO. Besides similar body weights between both groups, mIndy-ASO treatment lead to a 74% reduction in fasting plasma insulin concentrations as well as a 35% reduction in plasma triglycerides. Moreover, hepatic triglyceride content was significantly reduced by the knockdown of mIndy, likely mediating a trend to decreased basal rates of endogenous glucose production as well as an increased suppression of hepatic glucose production by 25% during a hyperinsulinemic-euglycemic clamp. Together, these data suggest that inducible liver-selective reduction of mIndy in rats is able to ameliorate hepatic steatosis and insulin resistance, conditions occurring with high calorie diets and during aging.
Collapse
Affiliation(s)
- Dominik H Pesta
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.,Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.,Department of Sport Science, Medical Section, University of Innsbruck, Innsbruck, Austria.,Department of Visceral, Transplant, and Thoracic Surgery, D. Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University Düsseldorf, German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Rachel J Perry
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.,Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.,Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | | | - Dongyan Zhang
- Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Michael Jurczak
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Antje Fischer-Rosinsky
- Charité - University School of Medicine, Department of Endocrinology, Diabetes and Nutrition, Berlin, Germany
| | - Martin A Daniels
- Charité - University School of Medicine, Department of Endocrinology, Diabetes and Nutrition, Berlin, Germany.,Section of Metabolic Vascular Medicine, Medical Clinic III and Paul Langerhans Institute Dresden (PLID), TU Dresden, Germany
| | - Diana M Willmes
- Section of Metabolic Vascular Medicine, Medical Clinic III and Paul Langerhans Institute Dresden (PLID), TU Dresden, Germany.,German Center for Diabetes Research (DZD), Dresden, Germany
| | | | - Stefan R Bornstein
- Section of Metabolic Vascular Medicine, Medical Clinic III and Paul Langerhans Institute Dresden (PLID), TU Dresden, Germany.,German Center for Diabetes Research (DZD), Dresden, Germany.,Section of Diabetes and Nutritional Sciences, Rayne Institute, King's College London, London, UK
| | - Felix Knauf
- University Clinic Erlangen, Erlangen, Germany
| | - Varman T Samuel
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.,Veterans Affairs Medical Center, West Haven, CT, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.,Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.,Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Andreas L Birkenfeld
- Section of Metabolic Vascular Medicine, Medical Clinic III and Paul Langerhans Institute Dresden (PLID), TU Dresden, Germany.,German Center for Diabetes Research (DZD), Dresden, Germany.,Section of Diabetes and Nutritional Sciences, Rayne Institute, King's College London, London, UK
| |
Collapse
|
14
|
Rogers RP, Rogina B. The role of INDY in metabolism, health and longevity. Front Genet 2015; 6:204. [PMID: 26106407 PMCID: PMC4460575 DOI: 10.3389/fgene.2015.00204] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Accepted: 05/25/2015] [Indexed: 11/24/2022] Open
Abstract
Indy (I’m Not Dead Yet) encodes the fly homolog of a mammalian SLC13A5 plasma membrane transporter. INDY is expressed in metabolically active tissues functioning as a transporter of Krebs cycle intermediates with the highest affinity for citrate. Decreased expression of the Indy gene extends longevity in Drosophila and C. elegans. Reduction of INDY or its respective homologs in C. elegans and mice induces metabolic and physiological changes similar to those observed in calorie restriction. It is thought that these physiological changes are due to altered levels of cytoplasmic citrate, which directly impacts Krebs cycle energy production as a result of shifts in substrate availability. Citrate cleavage is a key event during lipid and glucose metabolism; thus, reduction of citrate due to Indy reduction alters these processes. With regards to mammals, mice with reduced Indy (mIndy–/–) also exhibit changes in glucose metabolism, mitochondrial biogenesis and are protected from the negative effects of a high calorie diet. Together, these data support a role for Indy as a metabolic regulator, which suggests INDY as a therapeutic target for treatment of diet and age-related disorders such as Type II Diabetes and obesity.
Collapse
Affiliation(s)
- Ryan P Rogers
- Department of Sciences, Wentworth Institute of Technology , Boston, MA, USA
| | - Blanka Rogina
- Department of Genetics and Genome Sciences, Institute for Systems Genomics, School of Medicine, University of Connecticut Health Center , Farmington, CT, USA
| |
Collapse
|
15
|
Li L, Li H, Garzel B, Yang H, Sueyoshi T, Li Q, Shu Y, Zhang J, Hu B, Heyward S, Moeller T, Xie W, Negishi M, Wang H. SLC13A5 is a novel transcriptional target of the pregnane X receptor and sensitizes drug-induced steatosis in human liver. Mol Pharmacol 2015; 87:674-82. [PMID: 25628225 PMCID: PMC4366797 DOI: 10.1124/mol.114.097287] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 01/26/2015] [Indexed: 12/20/2022] Open
Abstract
The solute carrier family 13 member 5 (SLC13A5) is a sodium-coupled transporter that mediates cellular uptake of citrate, which plays important roles in the synthesis of fatty acids and cholesterol. Recently, the pregnane X receptor (PXR, NR1I2), initially characterized as a xenobiotic sensor, has been functionally linked to the regulation of various physiologic processes that are associated with lipid metabolism and energy homeostasis. Here, we show that the SLC13A5 gene is a novel transcriptional target of PXR, and altered expression of SLC13A5 affects lipid accumulation in human liver cells. The prototypical PXR activator rifampicin markedly induced the mRNA and protein expression of SLC13A5 in human primary hepatocytes. Utilizing cell-based luciferase reporter assays, electrophoretic mobility shift assays, and chromatin immunoprecipitation assays, we identified and functionally characterized two enhancer modules located upstream of the SLC13A5 gene transcription start site that are associated with regulation of PXR-mediated SLC13A5 induction. Functional analysis further revealed that rifampicin can enhance lipid accumulation in human primary hepatocytes, and knockdown of SLC13A5 expression alone leads to a significant decrease of the lipid content in HepG2 cells. Overall, our results uncover SLC13A5 as a novel target gene of PXR and may contribute to drug-induced steatosis and metabolic disorders in humans.
Collapse
Affiliation(s)
- Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Haishan Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Brandy Garzel
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Hui Yang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Tatsuya Sueyoshi
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Qing Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Yan Shu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Junran Zhang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Bingfang Hu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Scott Heyward
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Timothy Moeller
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Wen Xie
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Masahiko Negishi
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (L.L., H.L., B.G., H.Y., Q.L., Y.S., H.W.); Pharmacogenetics Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental and Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina (T.S., M.N.); Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio (J.Z.); Bioreclamation In Vitro Technologies, Baltimore, Maryland (S.H., T.M.); and Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (B.H., W.X.)
| |
Collapse
|
16
|
Gopal E, Babu E, Ramachandran S, Bhutia YD, Prasad PD, Ganapathy V. Species-specific influence of lithium on the activity of SLC13A5 (NaCT): lithium-induced activation is specific for the transporter in primates. J Pharmacol Exp Ther 2015; 353:17-26. [PMID: 25617245 DOI: 10.1124/jpet.114.221523] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
NaCT (SLC13A5) is a Na(+)-coupled transporter for Krebs cycle intermediates and is expressed predominantly in the liver. Human NaCT is relatively specific for citrate compared with other Krebs cycle intermediates. The transport activity of human NaCT is stimulated by Li(+), whereas that of rat NaCT is inhibited by Li(+). We studied the influence of Li(+) on NaCTs cloned from eight different species. Li(+) stimulated the activity of only NaCTs from primates (human, chimpanzee, and monkey); by contrast, NaCTs from nonprimate species (mouse, rat, dog, and zebrafish) were inhibited by Li(+). Caenorhabditis elegans NaCT was not affected by Li(+). With human NaCT, the Li(+)-induced increase in transport activity was associated with the conversion of the transporter from a low-affinity/high-capacity type to a high-affinity/low-capacity type. H(+) was able to substitute for Li(+) in eliciting the stimulatory effect. The amino acid Phe500 in human NaCT was critical for Li(+)/H(+)-induced stimulation. Mutation of this amino acid to tryptophan (F500W) markedly increased the basal transport activity of human NaCT in the absence of Li(+), but the ability of Li(+) to stimulate the transporter was almost completely lost with this mutant. Substitution of Phe500 with tryptophan in human NaCT converted the transporter from a low-affinity/high-capacity type to a high-affinity/low-capacity type, an effect similar to that of Li(+) on the wild-type NaCT. These studies show that Li(+)-induced activation of NaCT is specific for the transporter in primates and that the region surrounding Phe500 in primate NaCTs is important for the Li(+) effect.
Collapse
Affiliation(s)
- Elangovan Gopal
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas (E.B., S.R., Y.D.B., V.G.); and Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia (E.G., P.D.P.)
| | - Ellappan Babu
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas (E.B., S.R., Y.D.B., V.G.); and Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia (E.G., P.D.P.)
| | - Sabarish Ramachandran
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas (E.B., S.R., Y.D.B., V.G.); and Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia (E.G., P.D.P.)
| | - Yangzom D Bhutia
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas (E.B., S.R., Y.D.B., V.G.); and Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia (E.G., P.D.P.)
| | - Puttur D Prasad
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas (E.B., S.R., Y.D.B., V.G.); and Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia (E.G., P.D.P.)
| | - Vadivel Ganapathy
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas (E.B., S.R., Y.D.B., V.G.); and Department of Biochemistry and Molecular Biology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia (E.G., P.D.P.)
| |
Collapse
|
17
|
Rogers RP, Rogina B. Increased mitochondrial biogenesis preserves intestinal stem cell homeostasis and contributes to longevity in Indy mutant flies. Aging (Albany NY) 2014; 6:335-50. [PMID: 24827528 PMCID: PMC4032799 DOI: 10.18632/aging.100658] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The Drosophila Indy (I'm Not Dead Yet) gene encodes a plasma membrane transporter of Krebs cycle intermediates, with robust expression in tissues associated with metabolism. Reduced INDY alters metabolism and extends longevity in a manner similar to caloric restriction (CR); however, little is known about the tissue specific physiological effects of INDY reduction. Here we focused on the effects of INDY reduction in the Drosophila midgut due to the importance of intestinal tissue homeostasis in healthy aging and longevity. The expression of Indy mRNA in the midgut changes in response to aging and nutrition. Genetic reduction of Indy expression increases midgut expression of the mitochondrial regulator spargel/dPGC-1, which is accompanied by increased mitochondrial biogenesis and reduced reactive oxygen species (ROS). These physiological changes in the Indy mutant midgut preserve intestinal stem cell (ISC) homeostasis and are associated with healthy aging. Genetic studies confirm that dPGC-1 mediates the regulatory effects of INDY, as illustrated by lack of longevity extension and ISC homeostasis in flies with mutations in both Indy and dPGC1. Our data suggest INDY may be a physiological regulator that modulates intermediary metabolism in response to changes in nutrient availability and organismal needs by modulating dPGC-1
Collapse
Affiliation(s)
- Ryan P Rogers
- Department of Genetics and Developmental Biology, School of Medicine, University of Connecticut Health Center, 263 Farmington, CT 06030-6403, USA
| | | |
Collapse
|
18
|
Ma Y, Bai XY, Du X, Fu B, Chen X. NaDC3 Induces Premature Cellular Senescence by Promoting Transport of Krebs Cycle Intermediates, Increasing NADH, and Exacerbating Oxidative Damage. J Gerontol A Biol Sci Med Sci 2014; 71:1-12. [PMID: 25384549 DOI: 10.1093/gerona/glu198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 10/05/2014] [Indexed: 11/12/2022] Open
Abstract
High-affinity sodium-dependent dicarboxylate cotransporter 3 (NaDC3) is a key metabolism-regulating membrane protein responsible for transport of Krebs cycle intermediates. NaDC3 is upregulated as organs age, but knowledge regarding the underlying mechanisms by which NaDC3 modulates mammalian aging is limited. In this study, we showed that NaDC3 overexpression accelerated cellular senescence in young human diploid cells (MRC-5 and WI-38) and primary renal tubular cells, leading to cell cycle arrest in G1 phase and increased expression of senescent biomarkers, senescence-associated β-galactosidase and p16. Intracellular levels of reactive oxygen species, 8-hydroxy-2'-deoxyguanosine, malondialdehyde, and carbonyl were significantly enhanced, and activities of respiratory complexes I and III and ATP level were significantly decreased in NaDC3-infected cells. Stressful premature senescent phenotypes induced by NaDC3 were markedly ameliorated via treatment with the antioxidants Tiron and Tempol. High expression of NaDC3 caused a prominent increase in intracellular levels of Krebs cycle intermediates and NADH. Exogenous NADH and NAD(+) may aggravate and attenuate the aging phenotypes induced by NaDC3, respectively. These results suggest that NaDC3 can induce premature cellular senescence by promoting the transport of Krebs cycle intermediates, increasing generation of NADH and reactive oxygen species and leading to oxidative damage. Our results clarify the aging signaling pathway regulated by NaDC3.
Collapse
Affiliation(s)
- Yuxiang Ma
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China. Department of Internal Medicine, Beijing Chuiyangliu Hospital, China
| | - Xue-Yuan Bai
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Xuan Du
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Bo Fu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, China
| |
Collapse
|
19
|
DeSalvo MK, Hindle SJ, Rusan ZM, Orng S, Eddison M, Halliwill K, Bainton RJ. The Drosophila surface glia transcriptome: evolutionary conserved blood-brain barrier processes. Front Neurosci 2014; 8:346. [PMID: 25426014 PMCID: PMC4224204 DOI: 10.3389/fnins.2014.00346] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/10/2014] [Indexed: 12/29/2022] Open
Abstract
Central nervous system (CNS) function is dependent on the stringent regulation of metabolites, drugs, cells, and pathogens exposed to the CNS space. Cellular blood-brain barrier (BBB) structures are highly specific checkpoints governing entry and exit of all small molecules to and from the brain interstitial space, but the precise mechanisms that regulate the BBB are not well understood. In addition, the BBB has long been a challenging obstacle to the pharmacologic treatment of CNS diseases; thus model systems that can parse the functions of the BBB are highly desirable. In this study, we sought to define the transcriptome of the adult Drosophila melanogaster BBB by isolating the BBB surface glia with fluorescence activated cell sorting (FACS) and profiling their gene expression with microarrays. By comparing the transcriptome of these surface glia to that of all brain glia, brain neurons, and whole brains, we present a catalog of transcripts that are selectively enriched at the Drosophila BBB. We found that the fly surface glia show high expression of many ATP-binding cassette (ABC) and solute carrier (SLC) transporters, cell adhesion molecules, metabolic enzymes, signaling molecules, and components of xenobiotic metabolism pathways. Using gene sequence-based alignments, we compare the Drosophila and Murine BBB transcriptomes and discover many shared chemoprotective and small molecule control pathways, thus affirming the relevance of invertebrate models for studying evolutionary conserved BBB properties. The Drosophila BBB transcriptome is valuable to vertebrate and insect biologists alike as a resource for studying proteins underlying diffusion barrier development and maintenance, glial biology, and regulation of drug transport at tissue barriers.
Collapse
Affiliation(s)
- Michael K DeSalvo
- Department of Anesthesia and Perioperative Care, University of California San Francisco San Francisco, CA, USA
| | - Samantha J Hindle
- Department of Anesthesia and Perioperative Care, University of California San Francisco San Francisco, CA, USA
| | - Zeid M Rusan
- Department of Anesthesia and Perioperative Care, University of California San Francisco San Francisco, CA, USA
| | - Souvinh Orng
- Department of Anesthesia and Perioperative Care, University of California San Francisco San Francisco, CA, USA
| | - Mark Eddison
- Janelia Farm Research Campus, The Howard Hughes Medical Institute Ashburn, VA, USA
| | - Kyle Halliwill
- Pharmaceutical Sciences and Pharmacogenomics, University of California San Francisco San Francisco, CA, USA
| | - Roland J Bainton
- Department of Anesthesia and Perioperative Care, University of California San Francisco San Francisco, CA, USA
| |
Collapse
|
20
|
Neuschäfer-Rube F, Lieske S, Kuna M, Henkel J, Perry RJ, Erion DM, Pesta D, Willmes DM, Brachs S, von Loeffelholz C, Tolkachov A, Schupp M, Pathe-Neuschäfer-Rube A, Pfeiffer AF, Shulman GI, Püschel GP, Birkenfeld AL. The mammalian INDY homolog is induced by CREB in a rat model of type 2 diabetes. Diabetes 2014; 63:1048-57. [PMID: 24222346 PMCID: PMC3968437 DOI: 10.2337/db13-0749] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Reduced expression of the INDY (I'm not dead yet) tricarboxylate carrier increased the life span in different species by mechanisms akin to caloric restriction. Mammalian INDY homolog (mIndy, SLC13A5) gene expression seems to be regulated by hormonal and/or nutritional factors. The underlying mechanisms are still unknown. The current study revealed that mIndy expression and [(14)C]-citrate uptake was induced by physiological concentrations of glucagon via a cAMP-dependent and cAMP-responsive element-binding protein (CREB)-dependent mechanism in primary rat hepatocytes. The promoter sequence of mIndy located upstream of the most frequent transcription start site was determined by 5'-rapid amplification of cDNA ends. In silico analysis identified a CREB-binding site within this promoter fragment of mIndy. Functional relevance for the CREB-binding site was demonstrated with reporter gene constructs that were induced by CREB activation when under the control of a fragment of a wild-type promoter, whereas promoter activity was lost after site-directed mutagenesis of the CREB-binding site. Moreover, CREB binding to this promoter element was confirmed by chromatin immunoprecipitation in rat liver. In vivo studies revealed that mIndy was induced in livers of fasted as well as in high-fat-diet-streptozotocin diabetic rats, in which CREB is constitutively activated. mIndy induction was completely prevented when CREB was depleted in these rats by antisense oligonucleotides. Together, these data suggest that mIndy is a CREB-dependent glucagon target gene that is induced in fasting and in type 2 diabetes. Increased mIndy expression might contribute to the metabolic consequences of diabetes in the liver.
Collapse
Affiliation(s)
- Frank Neuschäfer-Rube
- University of Potsdam, Institute of Nutritional Science, Nutritional Biochemistry, Potsdam, Germany
| | - Stefanie Lieske
- University of Potsdam, Institute of Nutritional Science, Nutritional Biochemistry, Potsdam, Germany
- Charité–University School of Medicine, Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Berlin, Germany
| | - Manuela Kuna
- University of Potsdam, Institute of Nutritional Science, Nutritional Biochemistry, Potsdam, Germany
| | - Janin Henkel
- University of Potsdam, Institute of Nutritional Science, Nutritional Biochemistry, Potsdam, Germany
| | - Rachel J. Perry
- Howard Hughes Medical Institute and the Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT
| | - Derek M. Erion
- Howard Hughes Medical Institute and the Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT
- Cardiovascular, Metabolic and Endocrine Diseases Research Unit, Pfizer, Inc., Cambridge, MA
| | - Dominik Pesta
- Howard Hughes Medical Institute and the Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT
| | - Diana M. Willmes
- Charité–University School of Medicine, Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Berlin, Germany
| | - Sebastian Brachs
- Charité–University School of Medicine, Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Berlin, Germany
| | - Christian von Loeffelholz
- Department of Anesthesiology and Intensive Care, and Treatment Center, Center for Sepsis Control and Care, Jena University Hospital, Friedrich Schiller University of Jena, Jena, Germany
| | - Alexander Tolkachov
- Charité–University School of Medicine, Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Berlin, Germany
| | - Michael Schupp
- Charité–University School of Medicine, Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Berlin, Germany
| | | | - Andreas F.H. Pfeiffer
- Charité–University School of Medicine, Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Berlin, Germany
- German Institute of Human Nutrition Potsdam Rehbrücke, Department of Clinical Nutrition, Nuthetal, Germany
| | - Gerald I. Shulman
- Howard Hughes Medical Institute and the Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT
| | - Gerhard P. Püschel
- University of Potsdam, Institute of Nutritional Science, Nutritional Biochemistry, Potsdam, Germany
- Corresponding author: Gerhard P. Püschel,
| | - Andreas L. Birkenfeld
- Charité–University School of Medicine, Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Berlin, Germany
| |
Collapse
|
21
|
Willmes DM, Birkenfeld AL. The Role of INDY in Metabolic Regulation. Comput Struct Biotechnol J 2013; 6:e201303020. [PMID: 24688728 PMCID: PMC3962103 DOI: 10.5936/csbj.201303020] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 12/02/2013] [Accepted: 12/02/2013] [Indexed: 01/20/2023] Open
Abstract
Reduced expression of the Indy (I'm Not Dead Yet) gene in D. melanogaster and C. elegans extends longevity. Indy and its mammalian homolog mINDY (Slc13a5, NaCT) are transporters of TCA cycle intermediates, mainly handling the uptake of citrate via the plasma membrane into the cytosol. Deletion of mINDY in mice leads to significant metabolic changes akin to caloric restriction, likely caused by reducing the effects of mINDY-imported citrate on fatty acid and cholesterol synthesis, glucose metabolism and ß-oxidation. This review will provide an overview on different mammalian SLC1 3 family members with a focus on mINDY (SLCl3A5) in glucose and energy metabolism and will highlight the role of mINDY as a putative therapeutic target for the treatment of obesity, non-alcoholic fatty liver disease and type 2 diabetes.
Collapse
Affiliation(s)
- Diana M Willmes
- Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Charité - University School of Medicine, Berlin, Germany
| | - Andreas L Birkenfeld
- Department of Endocrinology, Diabetes and Nutrition, Center for Cardiovascular Research, Charité - University School of Medicine, Berlin, Germany
| |
Collapse
|
22
|
Sodium-coupled dicarboxylate and citrate transporters from the SLC13 family. Pflugers Arch 2013; 466:119-30. [PMID: 24114175 DOI: 10.1007/s00424-013-1369-y] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 09/19/2013] [Accepted: 09/23/2013] [Indexed: 12/30/2022]
Abstract
The SLC13 family in humans and other mammals consists of sodium-coupled transporters for anionic substrates: three transporters for dicarboxylates/citrate and two transporters for sulfate. This review will focus on the di- and tricarboxylate transporters: NaDC1 (SLC13A2), NaDC3 (SLC13A3), and NaCT (SLC13A5). The substrates of these transporters are metabolic intermediates of the citric acid cycle, including citrate, succinate, and α-ketoglutarate, which can exert signaling effects through specific receptors or can affect metabolic enzymes directly. The SLC13 transporters are important for regulating plasma, urinary and tissue levels of these metabolites. NaDC1, primarily found on the apical membranes of renal proximal tubule and small intestinal cells, is involved in regulating urinary levels of citrate and plays a role in kidney stone development. NaDC3 has a wider tissue distribution and high substrate affinity compared with NaDC1. NaDC3 participates in drug and xenobiotic excretion through interactions with organic anion transporters. NaCT is primarily a citrate transporter located in the liver and brain, and its activity may regulate metabolic processes. The recent crystal structure of the Vibrio cholerae homolog, VcINDY, provides a new framework for understanding the mechanism of transport in this family. This review summarizes current knowledge of the structure, function, and regulation of the di- and tricarboxylate transporters of the SLC13 family.
Collapse
|
23
|
The p66Shc gene paves the way for healthspan: Evolutionary and mechanistic perspectives. Neurosci Biobehav Rev 2013; 37:790-802. [DOI: 10.1016/j.neubiorev.2013.03.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Revised: 03/04/2013] [Accepted: 03/11/2013] [Indexed: 12/23/2022]
|
24
|
Abstract
Decreased expression of the fly and worm Indy genes extends longevity. The fly Indy gene and its mammalian homolog are transporters of Krebs cycle intermediates, with the highest rate of uptake for citrate. Cytosolic citrate has a role in energy regulation by affecting fatty acid synthesis and glycolysis. Fly, worm, and mice Indy gene homologs are predominantly expressed in places important for intermediary metabolism. Consequently, decreased expression of Indy in fly and worm, and the removal of mIndy in mice exhibit changes associated with calorie restriction, such as decreased levels of lipids, changes in carbohydrate metabolism and increased mitochondrial biogenesis. Here we report that several Indy alleles in a diverse array of genetic backgrounds confer increased longevity.
Collapse
Affiliation(s)
- Blanka Rogina
- Department of Genetics and Developmental Biology, School of Medicine, University of Connecticut Health Center Farmington, CT, USA
| | | |
Collapse
|
25
|
Bergeron M, Clémençon B, Hediger M, Markovich D. SLC13 family of Na+-coupled di- and tri-carboxylate/sulfate transporters. Mol Aspects Med 2013; 34:299-312. [DOI: 10.1016/j.mam.2012.12.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 11/16/2012] [Indexed: 12/22/2022]
|
26
|
Edwards CB, Copes N, Brito AG, Canfield J, Bradshaw PC. Malate and fumarate extend lifespan in Caenorhabditis elegans. PLoS One 2013; 8:e58345. [PMID: 23472183 PMCID: PMC3589421 DOI: 10.1371/journal.pone.0058345] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 02/03/2013] [Indexed: 11/18/2022] Open
Abstract
Malate, the tricarboxylic acid (TCA) cycle metabolite, increased lifespan and thermotolerance in the nematode C. elegans. Malate can be synthesized from fumarate by the enzyme fumarase and further oxidized to oxaloacetate by malate dehydrogenase with the accompanying reduction of NAD. Addition of fumarate also extended lifespan, but succinate addition did not, although all three intermediates activated nuclear translocation of the cytoprotective DAF-16/FOXO transcription factor and protected from paraquat-induced oxidative stress. The glyoxylate shunt, an anabolic pathway linked to lifespan extension in C. elegans, reversibly converts isocitrate and acetyl-CoA to succinate, malate, and CoA. The increased longevity provided by malate addition did not occur in fumarase (fum-1), glyoxylate shunt (gei-7), succinate dehydrogenase flavoprotein (sdha-2), or soluble fumarate reductase F48E8.3 RNAi knockdown worms. Therefore, to increase lifespan, malate must be first converted to fumarate, then fumarate must be reduced to succinate by soluble fumarate reductase and the mitochondrial electron transport chain complex II. Reduction of fumarate to succinate is coupled with the oxidation of FADH2 to FAD. Lifespan extension induced by malate depended upon the longevity regulators DAF-16 and SIR-2.1. Malate supplementation did not extend the lifespan of long-lived eat-2 mutant worms, a model of dietary restriction. Malate and fumarate addition increased oxygen consumption, but decreased ATP levels and mitochondrial membrane potential suggesting a mild uncoupling of oxidative phosphorylation. Malate also increased NADPH, NAD, and the NAD/NADH ratio. Fumarate reduction, glyoxylate shunt activity, and mild mitochondrial uncoupling likely contribute to the lifespan extension induced by malate and fumarate by increasing the amount of oxidized NAD and FAD cofactors.
Collapse
Affiliation(s)
- Clare B. Edwards
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida Tampa, Florida, United States of America
| | - Neil Copes
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida Tampa, Florida, United States of America
| | - Andres G. Brito
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida Tampa, Florida, United States of America
| | - John Canfield
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida Tampa, Florida, United States of America
| | - Patrick C. Bradshaw
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida Tampa, Florida, United States of America
- * E-mail:
| |
Collapse
|
27
|
Abstract
Indy encodes the fly homolog of a mammalian transporter of di and tricarboxylate components of the Krebs cycle. Reduced expression of fly Indy or two of the C. elegans Indy homologs leads to an increase in life span. Fly and worm tissues that play key roles in intermediary metabolism are also the places where Indy genes are expressed. One of the mouse homologs of Indy (mIndy) is mainly expressed in the liver. It has been hypothesized that decreased INDY activity creates a state similar to caloric restriction (CR). This hypothesis is supported by the physiological similarities between Indy mutant flies on high calorie food and control flies on CR, such as increased physical activity and decreases in weight, egg production, triglyceride levels, starvation resistance, and insulin signaling. In addition, Indy mutant flies undergo changes in mitochondrial biogenesis also observed in CR animals. Recent findings with mIndy knockout mice support and extend the findings from flies. mIndy−/− mice display an increase in hepatic mitochondrial biogenesis, lipid oxidation, and decreased hepatic lipogenesis. When mIndy−/− mice are fed high calorie food they are protected from adiposity and insulin resistance. These findings point to INDY as a potential drug target for the treatment of metabolic syndrome, type 2 diabetes, and obesity.
Collapse
Affiliation(s)
- Stewart Frankel
- Department of Biology, University of Hartford West Hartford, CT, USA
| | | |
Collapse
|
28
|
Birkenfeld AL, Lee HY, Guebre-Egziabher F, Alves TC, Jurczak MJ, Jornayvaz FR, Zhang D, Hsiao JJ, Martin-Montalvo A, Fischer-Rosinsky A, Spranger J, Pfeiffer AF, Jordan J, Fromm MF, König J, Lieske S, Carmean CM, Frederick DW, Weismann D, Knauf F, Irusta PM, De Cabo R, Helfand SL, Samuel VT, Shulman GI. Deletion of the mammalian INDY homolog mimics aspects of dietary restriction and protects against adiposity and insulin resistance in mice. Cell Metab 2011; 14:184-95. [PMID: 21803289 PMCID: PMC3163140 DOI: 10.1016/j.cmet.2011.06.009] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2010] [Revised: 12/24/2010] [Accepted: 06/07/2011] [Indexed: 01/07/2023]
Abstract
Reduced expression of the Indy (I'm Not Dead, Yet) gene in D. melanogaster and its homolog in C. elegans prolongs life span and in D. melanogaster augments mitochondrial biogenesis in a manner akin to caloric restriction. However, the cellular mechanism by which Indy does this is unknown. Here, we report on the knockout mouse model of the mammalian Indy (mIndy) homolog, SLC13A5. Deletion of mIndy in mice (mINDY(-/-) mice) reduces hepatocellular ATP/ADP ratio, activates hepatic AMPK, induces PGC-1α, inhibits ACC-2, and reduces SREBP-1c levels. This signaling network promotes hepatic mitochondrial biogenesis, lipid oxidation, and energy expenditure and attenuates hepatic de novo lipogenesis. Together, these traits protect mINDY(-/-) mice from the adiposity and insulin resistance that evolve with high-fat feeding and aging. Our studies demonstrate a profound effect of mIndy on mammalian energy metabolism and suggest that mINDY might be a therapeutic target for the treatment of obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Andreas L Birkenfeld
- Howard Hughes Medical Institute, Yale School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Schachter H, Boulianne G. Life is sweet! A novel role for N-glycans in Drosophila lifespan. Fly (Austin) 2011; 5:18-24. [PMID: 21057214 DOI: 10.4161/fly.5.1.13920] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
N-glycans are post-translational modifications in which the sugar chain is covalently linked to protein by a GlcNAcβ1-N-asparagine linkage. Drosophila melanogaster and other invertebrates, but not vertebrates, synthesize large amounts of "paucimannose" N-glycans that contain only three or four mannose residues. The enzyme UDP-GlcNAc:α3-D-mannoside β1,2-N-acetylglucosaminyltransferase I (GnTI, encoded by the Mgat1 gene) controls the synthesis of paucimannose N-glycans. Either deletion or neuron-specific knockdown of Mgat1 in wild type flies results in pronounced defects in locomotion, structural defects in the adult central nervous system and a severely reduced lifespan. We have recently shown that neuronal expression of a wild-type Mgat1 transgene in Mgat1-null flies rescues the structural defects in the brain (fused β-lobes) and the shortened lifespan and, surprisingly, results in a dramatic 135% increase in mean lifespan relative to genetically identical controls that do not express the transgene. In this review, we discuss various approaches that can be used to determine the roles of paucimannose N-glycans in Drosophila longevity and in the adult CNS.
Collapse
Affiliation(s)
- Harry Schachter
- Program in Molecular Structure and Function, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada.
| | | |
Collapse
|
30
|
Sun J, Aluvila S, Kotaria R, Mayor JA, Walters DE, Kaplan RS. Mitochondrial and Plasma Membrane Citrate Transporters: Discovery of Selective Inhibitors and Application to Structure/Function Analysis. MOLECULAR AND CELLULAR PHARMACOLOGY 2010; 2:101-110. [PMID: 20686672 PMCID: PMC2913483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Cytoplasmic citrate is the prime carbon source for fatty acid, triacylglycerol, and cholesterol biosyntheses, and also regulates glucose metabolism via its allosteric inhibition of phosphofructokinase. It originates either via the efflux of citrate from the mitochondrial matrix on the inner membrane citrate transport protein (CTP) or via the influx of extracellular citrate on the plasma membrane citrate transporter (PMCT). Despite their common substrate, the two transport proteins share little sequence similarity and they transport citrate via fundamentally different mechanisms. We tested the ability of a set of previously identified CTP inhibitors, to inhibit the PMCT. We found that of the top 10 CTP inhibitors only one substantially inhibited the PMCT. Conversely, we identified two other inhibitors that inhibited the PMCT but had little effect on the CTP. All three identified PMCT inhibitors displayed a noncompetitive mechanism. Furthermore, models to explain inhibitor interactions with the CTP are proposed. As part of the present studies a PMCT homology model has been developed based on the crystal structure of the leucine transporter, and a possible citrate binding site has been identified and its composition compared with the two known citrate binding sites present within the CTP. The ability to selectively inhibit the PMCT may prove key to the pharmacologic amelioration of metabolic disorders resulting from the synthesis of excess lipid, cholesterol, and glucose, including human obesity, hyperlipidemia, hyper-cholesterolemia, and Type 2 diabetes.
Collapse
Affiliation(s)
- Jiakang Sun
- Department of Biochemistry and Molecular Biology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | - Sreevidya Aluvila
- Department of Biochemistry and Molecular Biology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | - Rusudan Kotaria
- Department of Biochemistry and Molecular Biology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | - June A. Mayor
- Department of Biochemistry and Molecular Biology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | - D. Eric Walters
- Department of Biochemistry and Molecular Biology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | - Ronald S. Kaplan
- Department of Biochemistry and Molecular Biology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| |
Collapse
|
31
|
Aliverdieva DA, Mamaev DV. Molecular characteristics of transporters of C4-dicarboxylates and mechanism of translocation. J EVOL BIOCHEM PHYS+ 2009. [DOI: 10.1134/s0022093009030016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
32
|
Long-lived Indy and calorie restriction interact to extend life span. Proc Natl Acad Sci U S A 2009; 106:9262-7. [PMID: 19470468 DOI: 10.1073/pnas.0904115106] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Calorie restriction (CR) improves health and extends life span in a variety of species. Despite many downstream molecules and physiological systems having been identified as being regulated by CR, the mechanism by which CR extends life span remains unclear. The Drosophila gene Indy (for I'm not dead yet), involved in the transport and storage of Krebs cycle intermediates in tissues important in fly metabolism, was proposed to regulate life span via an effect on metabolism that could overlap with CR. In this study, we report that CR down regulates Indy mRNA expression, and that CR and the level of Indy expression interact to affect longevity. Optimal life span extension is seen when Indy expression is decreased between 25 and 75% of normal. Indy long-lived flies show several phenotypes that are shared by long-lived CR flies, including decreased insulin-like signaling, lipid storage, weight gain, and resistance to starvation as well as an increase in spontaneous physical activity. We conclude that Indy and CR interact to affect longevity and that a decrease in Indy may induce a CR-like status that confers life span extension.
Collapse
|
33
|
Abstract
Cells lacking aerobic metabolism because of damaged mtDNA accumulate in many postmitotic tissues in the course aging. Although being only a small fraction of cells, they might play a major role in oxidative stress affecting the whole body. However, it remains unclear how such cells, which are under normal circumstances dependent on aerobic metabolism, are able to survive for decades in vivo. Here a new model is presented that proposes a coexistence of anaerobic glycolysis and a partly reversed TCA cycle. Succinate plays a key role in the changed metabolic pathways because it has to be exported by the cell. This hypothesis supports the view that some respiration-deficient cells are able to survive permanently within the body and contribute to human aging.
Collapse
|
34
|
Toivonen JM, Walker GA, Martinez-Diaz P, Bjedov I, Driege Y, Jacobs HT, Gems D, Partridge L. No influence of Indy on lifespan in Drosophila after correction for genetic and cytoplasmic background effects. PLoS Genet 2007; 3:e95. [PMID: 17571923 PMCID: PMC1892600 DOI: 10.1371/journal.pgen.0030095] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2007] [Accepted: 04/26/2007] [Indexed: 01/10/2023] Open
Abstract
To investigate whether alterations in mitochondrial metabolism affect longevity in Drosophila melanogaster, we studied lifespan in various single gene mutants, using inbred and outbred genetic backgrounds. As positive controls we included the two most intensively studied mutants of Indy, which encodes a Drosophila Krebs cycle intermediate transporter. It has been reported that flies heterozygous for these Indy mutations, which lie outside the coding region, show almost a doubling of lifespan. We report that only one of the two mutants lowers mRNA levels, implying that the lifespan extension observed is not attributable to the Indy mutations themselves. Moreover, neither Indy mutation extended lifespan in female flies in any genetic background tested. In the original genetic background, only the Indy mutation associated with altered RNA expression extended lifespan in male flies. However, this effect was abolished by backcrossing into standard outbred genetic backgrounds, and was associated with an unidentified locus on the X chromosome. The original Indy line with long-lived males is infected by the cytoplasmic symbiont Wolbachia, and the longevity of Indy males disappeared after tetracycline clearance of this endosymbiont. These findings underscore the critical importance of standardisation of genetic background and of cytoplasm in genetic studies of lifespan, and show that the lifespan extension previously claimed for Indy mutants was entirely attributable to confounding variation from these two sources. In addition, we saw no effects on lifespan of expression knockdown of the Indy orthologues nac-2 and nac-3 in the nematode Caenorhabditis elegans.
Collapse
Affiliation(s)
- Janne M Toivonen
- Department of Biology, University College London, London, United Kingdom
- Institute of Medical Technology and Tampere University Hospital, FI-33014, University of Tampere, Tampere, Finland
| | - Glenda A Walker
- Department of Biology, University College London, London, United Kingdom
| | | | - Ivana Bjedov
- Department of Biology, University College London, London, United Kingdom
| | - Yasmine Driege
- Department of Biology, University College London, London, United Kingdom
| | - Howard T Jacobs
- Institute of Medical Technology and Tampere University Hospital, FI-33014, University of Tampere, Tampere, Finland
| | - David Gems
- Department of Biology, University College London, London, United Kingdom
| | - Linda Partridge
- Department of Biology, University College London, London, United Kingdom
| |
Collapse
|
35
|
Tong WH, Rouault TA. Metabolic regulation of citrate and iron by aconitases: role of iron–sulfur cluster biogenesis. Biometals 2007; 20:549-64. [PMID: 17205209 DOI: 10.1007/s10534-006-9047-6] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2006] [Accepted: 11/28/2006] [Indexed: 12/21/2022]
Abstract
Iron and citrate are essential for the metabolism of most organisms, and regulation of iron and citrate biology at both the cellular and systemic levels is critical for normal physiology and survival. Mitochondrial and cytosolic aconitases catalyze the interconversion of citrate and isocitrate, and aconitase activities are affected by iron levels, oxidative stress and by the status of the Fe-S cluster biogenesis apparatus. Assembly and disassembly of Fe-S clusters is a key process not only in regulating the enzymatic activity of mitochondrial aconitase in the citric acid cycle, but also in controlling the iron sensing and RNA binding activities of cytosolic aconitase (also known as iron regulatory protein IRP1). This review discusses the central role of aconitases in intermediary metabolism and explores how iron homeostasis and Fe-S cluster biogenesis regulate the Fe-S cluster switch and modulate intracellular citrate flux.
Collapse
Affiliation(s)
- Wing-Hang Tong
- Cell Biology and Metabolism Branch, National Institute of Child Health and Human Development, NIH Bldg 18, Rm 101, Bethesda, MD 20892, USA
| | | |
Collapse
|