151
|
Gorgoglione R, Porcelli V, Santoro A, Daddabbo L, Vozza A, Monné M, Di Noia MA, Palmieri L, Fiermonte G, Palmieri F. The human uncoupling proteins 5 and 6 (UCP5/SLC25A14 and UCP6/SLC25A30) transport sulfur oxyanions, phosphate and dicarboxylates. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2019; 1860:724-733. [PMID: 31356773 DOI: 10.1016/j.bbabio.2019.07.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/27/2019] [Accepted: 07/25/2019] [Indexed: 01/07/2023]
Abstract
The human genome encodes 53 members of the solute carrier family 25 (SLC25), also called the mitochondrial carrier family. In this work, two members of this family, UCP5 (BMCP1, brain mitochondrial carrier protein 1 encoded by SLC25A14) and UCP6 (KMCP1, kidney mitochondrial carrier protein 1 encoded by SLC25A30) have been thoroughly characterized biochemically. They were overexpressed in bacteria, purified and reconstituted in phospholipid vesicles. Their transport properties and kinetic parameters demonstrate that UCP5 and UCP6 transport inorganic anions (sulfate, sulfite, thiosulfate and phosphate) and, to a lesser extent, a variety of dicarboxylates (e.g. malonate, malate and citramalate) and, even more so, aspartate and (only UCP5) glutamate and tricarboxylates. Both carriers catalyzed a fast counter-exchange transport and a very low uniport of substrates. Transport was saturable and inhibited by mercurials and other mitochondrial carrier inhibitors at various degrees. The transport affinities of UCP5 and UCP6 were higher for sulfate and thiosulfate than for any other substrate, whereas the specific activity of UCP5 was much higher than that of UCP6. It is proposed that a main physiological role of UCP5 and UCP6 is to catalyze the export of sulfite and thiosulfate (the H2S degradation products) from the mitochondria, thereby modulating the level of the important signal molecule H2S.
Collapse
Affiliation(s)
- Ruggiero Gorgoglione
- Department of Biosciences, Biotechnologies and Biopharmaceutics, Laboratory of Biochemistry and Molecular Biology, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Vito Porcelli
- Department of Biosciences, Biotechnologies and Biopharmaceutics, Laboratory of Biochemistry and Molecular Biology, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Antonella Santoro
- Department of Biosciences, Biotechnologies and Biopharmaceutics, Laboratory of Biochemistry and Molecular Biology, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Lucia Daddabbo
- Department of Biosciences, Biotechnologies and Biopharmaceutics, Laboratory of Biochemistry and Molecular Biology, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Angelo Vozza
- Department of Biosciences, Biotechnologies and Biopharmaceutics, Laboratory of Biochemistry and Molecular Biology, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; Center of Excellence in Comparative Genomics, University of Bari, via Orabona 4, 70125 Bari, Italy
| | - Magnus Monné
- Department of Biosciences, Biotechnologies and Biopharmaceutics, Laboratory of Biochemistry and Molecular Biology, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; Department of Sciences, University of Basilicata, Via Ateneo Lucano 10, 85100 Potenza, Italy
| | - Maria Antonietta Di Noia
- Department of Biosciences, Biotechnologies and Biopharmaceutics, Laboratory of Biochemistry and Molecular Biology, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy
| | - Luigi Palmieri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, Laboratory of Biochemistry and Molecular Biology, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; Center of Excellence in Comparative Genomics, University of Bari, via Orabona 4, 70125 Bari, Italy; CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), 70126 Bari, Italy
| | - Giuseppe Fiermonte
- Department of Biosciences, Biotechnologies and Biopharmaceutics, Laboratory of Biochemistry and Molecular Biology, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; Center of Excellence in Comparative Genomics, University of Bari, via Orabona 4, 70125 Bari, Italy.
| | - Ferdinando Palmieri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, Laboratory of Biochemistry and Molecular Biology, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; Center of Excellence in Comparative Genomics, University of Bari, via Orabona 4, 70125 Bari, Italy; CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), 70126 Bari, Italy
| |
Collapse
|
152
|
Bertholet AM, Chouchani ET, Kazak L, Angelin A, Fedorenko A, Long JZ, Vidoni S, Garrity R, Cho J, Terada N, Wallace DC, Spiegelman BM, Kirichok Y. H + transport is an integral function of the mitochondrial ADP/ATP carrier. Nature 2019; 571:515-520. [PMID: 31341297 PMCID: PMC6662629 DOI: 10.1038/s41586-019-1400-3] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/23/2019] [Indexed: 12/22/2022]
Abstract
The mitochondrial ADP/ATP carrier (AAC) is a major transport protein of the inner mitochondrial membrane. It exchanges mitochondrial ATP for cytosolic ADP and controls cellular production of ATP. In addition, it has been proposed that AAC mediates mitochondrial uncoupling, but it has proven difficult to demonstrate this function or to elucidate its mechanisms. Here we record AAC currents directly from inner mitochondrial membranes from various mouse tissues and identify two distinct transport modes: ADP/ATP exchange and H+ transport. The AAC-mediated H+ current requires free fatty acids and resembles the H+ leak via the thermogenic uncoupling protein 1 found in brown fat. The ADP/ATP exchange via AAC negatively regulates the H+ leak, but does not completely inhibit it. This suggests that the H+ leak and mitochondrial uncoupling could be dynamically controlled by cellular ATP demand and the rate of ADP/ATP exchange. By mediating two distinct transport modes, ADP/ATP exchange and H+ leak, AAC connects coupled (ATP production) and uncoupled (thermogenesis) energy conversion in mitochondria.
Collapse
Affiliation(s)
- Ambre M Bertholet
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Edward T Chouchani
- Dana-Farber Cancer Institute & Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Lawrence Kazak
- Dana-Farber Cancer Institute & Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Alessia Angelin
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andriy Fedorenko
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Jonathan Z Long
- Dana-Farber Cancer Institute & Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Sara Vidoni
- Dana-Farber Cancer Institute & Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Ryan Garrity
- Dana-Farber Cancer Institute & Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Joonseok Cho
- Department of Pathology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Naohiro Terada
- Department of Pathology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bruce M Spiegelman
- Dana-Farber Cancer Institute & Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Yuriy Kirichok
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
153
|
Jesinkey SR, Madiraju AK, Alves TC, Yarborough OH, Cardone RL, Zhao X, Parsaei Y, Nasiri AR, Butrico G, Liu X, Molina AJ, Rountree AM, Neal AS, Wolf DM, Sterpka J, Philbrick WM, Sweet IR, Shirihai OH, Kibbey RG. Mitochondrial GTP Links Nutrient Sensing to β Cell Health, Mitochondrial Morphology, and Insulin Secretion Independent of OxPhos. Cell Rep 2019; 28:759-772.e10. [PMID: 31315053 PMCID: PMC6713209 DOI: 10.1016/j.celrep.2019.06.058] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 02/15/2019] [Accepted: 06/14/2019] [Indexed: 12/18/2022] Open
Abstract
Mechanisms coordinating pancreatic β cell metabolism with insulin secretion are essential for glucose homeostasis. One key mechanism of β cell nutrient sensing uses the mitochondrial GTP (mtGTP) cycle. In this cycle, mtGTP synthesized by succinyl-CoA synthetase (SCS) is hydrolyzed via mitochondrial PEPCK (PEPCK-M) to make phosphoenolpyruvate, a high-energy metabolite that integrates TCA cycling and anaplerosis with glucose-stimulated insulin secretion (GSIS). Several strategies, including xenotopic overexpression of yeast mitochondrial GTP/GDP exchanger (GGC1) and human ATP and GTP-specific SCS isoforms, demonstrated the importance of the mtGTP cycle. These studies confirmed that mtGTP triggers and amplifies normal GSIS and rescues defects in GSIS both in vitro and in vivo. Increased mtGTP synthesis enhanced calcium oscillations during GSIS. mtGTP also augmented mitochondrial mass, increased insulin granule number, and membrane proximity without triggering de-differentiation or metabolic fragility. These data highlight the importance of the mtGTP signal in nutrient sensing, insulin secretion, mitochondrial maintenance, and β cell health.
Collapse
Affiliation(s)
- Sean R Jesinkey
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Anila K Madiraju
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA; Departments of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Tiago C Alves
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - OrLando H Yarborough
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Rebecca L Cardone
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA; Departments of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Xiaojian Zhao
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Yassmin Parsaei
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Ali R Nasiri
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Gina Butrico
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Xinran Liu
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Anthony J Molina
- Division of Geriatrics and Gerontology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Austin M Rountree
- University of Washington Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Adam S Neal
- University of Washington Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Dane M Wolf
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA; Departments of Medicine, Endocrinology, and Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - John Sterpka
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - William M Philbrick
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Ian R Sweet
- University of Washington Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Orian H Shirihai
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA; Departments of Medicine, Endocrinology, and Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Richard G Kibbey
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06519, USA; Departments of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06519, USA.
| |
Collapse
|
154
|
Alkan HF, Bogner-Strauss JG. Maintaining cytosolic aspartate levels is a major function of the TCA cycle in proliferating cells. Mol Cell Oncol 2019; 6:e1536843. [PMID: 31528687 PMCID: PMC6736317 DOI: 10.1080/23723556.2018.1536843] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/12/2018] [Accepted: 10/13/2018] [Indexed: 01/16/2023]
Abstract
Cancer cells rely on glutamine to fuel mitochondria, however it remains unclear whether this is needed for bioenergetic or biosynthetic pathways. Our study suggests that an essential function of mitochondrial glutamine metabolism is to provide aspartate to the cytosol where it can be used for nucleotide and protein synthesis.
Collapse
Affiliation(s)
- H Furkan Alkan
- Institute of Biochemistry, Graz University of Technology, Graz, Austria
| | | |
Collapse
|
155
|
Ogando DG, Choi M, Shyam R, Li S, Bonanno JA. Ammonia sensitive SLC4A11 mitochondrial uncoupling reduces glutamine induced oxidative stress. Redox Biol 2019; 26:101260. [PMID: 31254733 PMCID: PMC6604051 DOI: 10.1016/j.redox.2019.101260] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/07/2019] [Accepted: 06/21/2019] [Indexed: 02/07/2023] Open
Abstract
SLC4A11 is a NH3 sensitive membrane transporter with H+ channel-like properties that facilitates Glutamine catabolism in Human and Mouse corneal endothelium (CE). Loss of SLC4A11 activity induces oxidative stress and cell death, resulting in Congenital Hereditary Endothelial Dystrophy (CHED) with corneal edema and vision loss. However, the mechanism by which SLC4A11 prevents ROS production and protects CE is unknown. Here we demonstrate that SLC4A11 is localized to the inner mitochondrial membrane of CE and SLC4A11 transfected PS120 fibroblasts, where it acts as an NH3-sensitive mitochondrial uncoupler that enhances glutamine-dependent oxygen consumption, electron transport chain activity, and ATP levels by suppressing damaging Reactive Oxygen Species (ROS) production. In the presence of glutamine, Slc4a11-/- (KO) mouse CE generate significantly greater mitochondrial superoxide, a greater proportion of damaged depolarized mitochondria, and more apoptotic cells than WT. KO CE can be rescued by MitoQ, reducing NH3 production by GLS1 inhibition or dimethyl αKetoglutarate supplementation, or by BAM15 mitochondrial uncoupling. Slc4a11 KO mouse corneal edema can be partially reversed by αKetoglutarate eye drops. Moreover, we demonstrate that this role for SLC4A11 is not specific to CE cells, as SLC4A11 knockdown in glutamine-addicted colon carcinoma cells reduced glutamine catabolism, increased ROS production, and inhibited cell proliferation. Overall, our studies reveal a unique metabolic mechanism that reduces mitochondrial oxidative stress while promoting glutamine catabolism.
Collapse
Affiliation(s)
- Diego G Ogando
- Indiana University, School of Optometry, Bloomington, IN, 47405, United States
| | - Moonjung Choi
- Indiana University, School of Optometry, Bloomington, IN, 47405, United States
| | - Rajalekshmy Shyam
- Indiana University, School of Optometry, Bloomington, IN, 47405, United States
| | - Shimin Li
- Indiana University, School of Optometry, Bloomington, IN, 47405, United States
| | - Joseph A Bonanno
- Indiana University, School of Optometry, Bloomington, IN, 47405, United States.
| |
Collapse
|
156
|
Mitochondrial Activity and Skeletal Muscle Insulin Resistance in Kidney Disease. Int J Mol Sci 2019; 20:ijms20112751. [PMID: 31195596 PMCID: PMC6600571 DOI: 10.3390/ijms20112751] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 05/28/2019] [Accepted: 06/04/2019] [Indexed: 12/17/2022] Open
Abstract
Insulin resistance is a key feature of the metabolic syndrome, a cluster of medical disorders that together increase the chance of developing type 2 diabetes and cardiovascular disease. In turn, type 2 diabetes may cause complications such as diabetic kidney disease (DKD). Obesity is a major risk factor for developing systemic insulin resistance, and skeletal muscle is the first tissue in susceptible individuals to lose its insulin responsiveness. Interestingly, lean individuals are not immune to insulin resistance either. Non-obese, non-diabetic subjects with chronic kidney disease (CKD), for example, exhibit insulin resistance at the very onset of CKD, even before clinical symptoms of renal failure are clear. This uraemic insulin resistance contributes to the muscle weakness and muscle wasting that many CKD patients face, especially during the later stages of the disease. Bioenergetic failure has been associated with the loss of skeletal muscle insulin sensitivity in obesity and uraemia, as well as in the development of kidney disease and its sarcopenic complications. In this mini review, we evaluate how mitochondrial activity of different renal cell types changes during DKD progression, and discuss the controversial role of oxidative stress and mitochondrial reactive oxygen species in DKD. We also compare the involvement of skeletal muscle mitochondria in uraemic and obesity-related muscle insulin resistance.
Collapse
|
157
|
Metabolic Phenotyping of Adipose-Derived Stem Cells Reveals a Unique Signature and Intrinsic Differences between Fat Pads. Stem Cells Int 2019; 2019:9323864. [PMID: 31223312 PMCID: PMC6541987 DOI: 10.1155/2019/9323864] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/11/2019] [Accepted: 02/03/2019] [Indexed: 12/20/2022] Open
Abstract
White adipose tissues are functionally heterogeneous and differently manage the excess of energy supply. While the expansion of subcutaneous adipose tissues (SAT) is protective in obesity, that of visceral adipose tissues (VAT) correlates with the emergence of metabolic diseases. Maintained in fat pads throughout life, adipose stem cells (ASC) are mesenchymal-like stem cells with adipogenesis and multipotent differentiation potential. ASC from distinct fat pads have long been reported to present distinct proliferation and differentiation potentials that are maintained in culture, yet the origins of these intrinsic differences are still unknown. Metabolism is central to stem cell fate decision in line with environmental changes. In this study, we performed high-resolution nuclear magnetic resonance (NMR) metabolomic analyses of ASC culture supernatants in order to characterize their metabolic phenotype in culture. We identified and quantified 29 ASC exometabolites and evaluated their consumption or secretion over 72 h of cell culture. Both ASC used glycolysis and mitochondrial metabolism, as evidenced by the high secretions of lactate and citrate, respectively, but V-ASC mostly used glycolysis. By varying the composition of the cell culture medium, we showed that glutaminolysis, rather than glycolysis, supported the secretion of pyruvate, alanine, and citrate, evidencing a peculiar metabolism in ASC cells. The comparison of the two types of ASC in glutamine-free culture conditions also revealed the role of glutaminolysis in the limitation of pyruvate routing towards the lactate synthesis, in S-ASC but not in V-ASC. Altogether, our results suggest a difference between depots in the capacity of ASC mitochondria to assimilate pyruvate, with probable consequences on their differentiation potential in pathways requiring an increased mitochondrial activity. These results highlight a pivotal role of metabolic mechanisms in the discrimination between ASC and provide new perspectives in the understanding of their functional differences.
Collapse
|
158
|
Pohl EE, Rupprecht A, Macher G, Hilse KE. Important Trends in UCP3 Investigation. Front Physiol 2019; 10:470. [PMID: 31133866 PMCID: PMC6524716 DOI: 10.3389/fphys.2019.00470] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 04/04/2019] [Indexed: 11/13/2022] Open
Abstract
Membrane uncoupling protein 3 (UCP3), a member of the mitochondrial uncoupling protein family, was discovered in 1997. UCP3's properties, such as its high homology to other mitochondrial carriers, especially to UCP2, its short lifetime and low specificity of UCP3 antibodies, have hindered progress in understanding its biological function and transport mechanism over decades. The abundance of UCP3 is highest in murine brown adipose tissue (BAT, 15.0 pmol/mg protein), compared to heart (2.7 pmol/mg protein) and the gastrocnemius muscle (1.7 pmol/mg protein), but it is still 400-fold lower than the abundance of UCP1, a biomarker for BAT. Investigation of UCP3 reconstituted in planar bilayer membranes revealed that it transports protons only when activated by fatty acids (FA). Although purine nucleotides (PN) inhibit UCP3-mediated transport, the molecular mechanism differs from that of UCP1. It remains a conundrum that two homologous proton-transporting proteins exist within the same tissue. Recently, we proposed that UCP3 abundance directly correlates with the degree of FA β-oxidation in cell metabolism. Further development in this field implies that UCP3 may have dual function in transporting substrates, which have yet to be identified, alongside protons. Evaluation of the literature with respect to UCP3 is a complex task because (i) UCP3 features are often extrapolated from its "twin" UCP2 without additional proof, and (ii) the specificity of antibodies against UCP3 used in studies is rarely evaluated. In this review, we primarily focus on recent findings obtained for UCP3 in biological and biomimetic systems.
Collapse
Affiliation(s)
- Elena E. Pohl
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Anne Rupprecht
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Rostock, Germany
| | - Gabriel Macher
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Karolina E. Hilse
- Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
159
|
Upregulation of UCP2 Expression Protects against LPS-Induced Oxidative Stress and Apoptosis in Cardiomyocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2758262. [PMID: 31182990 PMCID: PMC6512061 DOI: 10.1155/2019/2758262] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 01/21/2019] [Accepted: 03/10/2019] [Indexed: 12/17/2022]
Abstract
Uncoupling protein 2 (UCP2) has a cardioprotective role under septic conditions, but the underlying mechanism remains unclear. This study aimed at investigating the effects of UCP2 on the oxidative stress and apoptosis of cardiomyocytes induced by lipopolysaccharide (LPS). First, LPS increased UCP2 expression in cardiomyocytes in a time-dependent manner. LPS increased the production of lactate dehydrogenase (LDH), reactive oxygen species (ROS), and malondialdehyde (MDA) and decreased the level of superoxide dismutase (SOD). However, UCP2 knockdown increased the LPS-induced cardiac injury and oxidative stress. In addition, LPS damaged the mitochondrial ultrastructure and led to the disruption of mitochondrial membrane potential (MMP), as well as the release of mitochondrial cytochrome c. UCP2 knockdown aggravated mitochondrial injury and the release of mitochondrial cytochrome c. LPS increased the protein levels of Bax and cleaved-caspase-3, decreased the protein level of Bcl-2, and upregulated the protein level of mitogen-activated protein kinase. However, upon UCP2 knockdown, the protein levels of Bax and cleaved-caspase-3 increased even further, and the protein level of Bcl-2 was further decreased. The protein level of phosphorylated p38 was also further enhanced. Thus, UCP2 protects against LPS-induced oxidative stress and apoptosis in cardiomyocytes.
Collapse
|
160
|
Jazwinski SM, Kim S. Examination of the Dimensions of Biological Age. Front Genet 2019; 10:263. [PMID: 30972107 PMCID: PMC6445152 DOI: 10.3389/fgene.2019.00263] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/08/2019] [Indexed: 12/22/2022] Open
Abstract
The concept of biological age has been used more and more frequently in aging research in attempts to measure the progress of the biological aging process as opposed to the simple passage of time. Several approaches to quantify biological age have been utilized, including the use of biomarkers in the form of serum analytes, epigenetic markers, and deficit or frailty indices. Among these methods, the deficit index possesses a theoretical basis grounded in systems biology by incorporating networks, with their emergent properties, to describe the complex aging system. Application of the deficit index in human aging studies points to the increased energetic demands posed by an aging system that is losing integration. Different aspects of mitochondrial function appear to be responsible in males and females. The gut microbiome loses complexity in tandem with the host, as biological age increases, with likely impact on host metabolism and immunity. Specific DNA methylation changes are associated with biological age. They suggest declining connectivity within the aging network, at the cellular level. The deficit/frailty index may account for at least part of the departure at older ages of the observed mortality in the population from the exponential increase modeled by the Gompertz equation.
Collapse
Affiliation(s)
- S Michal Jazwinski
- Tulane Center for Aging, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA, United States
| | - Sangkyu Kim
- Tulane Center for Aging, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
161
|
Rupprecht A, Moldzio R, Mödl B, Pohl EE. Glutamine regulates mitochondrial uncoupling protein 2 to promote glutaminolysis in neuroblastoma cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2019; 1860:391-401. [PMID: 30885735 DOI: 10.1016/j.bbabio.2019.03.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/30/2018] [Accepted: 03/14/2019] [Indexed: 01/26/2023]
Abstract
Mitochondrial uncoupling protein 2 (UCP2) is highly abundant in rapidly proliferating cells that utilize aerobic glycolysis, such as stem cells, cancer cells, and cells of the immune system. However, the function of UCP2 has been a longstanding conundrum. Considering the strict regulation and unusually short life time of the protein, we propose that UCP2 acts as a "signaling protein" under nutrient shortage in cancer cells. We reveal that glutamine shortage induces the rapid and reversible downregulation of UCP2, decrease of the metabolic activity and proliferation of neuroblastoma cells, that are regulated by glutamine per se but not by glutamine metabolism. Our findings indicate a very rapid (within 1 h) metabolic adaptation that allows the cell to survive by either shifting its metabolism to the use of the alternative fuel glutamine or going into a reversible, more quiescent state. The results imply that UCP2 facilitates glutamine utilization as an energetic fuel source, thereby providing metabolic flexibility during glucose shortage. The targeting UCP2 by drugs to intervene with cancer cell metabolism may represent a new strategy for treatment of cancers resistant to other therapies.
Collapse
Affiliation(s)
- Anne Rupprecht
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria; Institute of Pharmacology and Toxicology, Rostock University Medical Center, Germany.
| | - Rudolf Moldzio
- Institute of Medical Biochemistry, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Bernadette Mödl
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Elena E Pohl
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria.
| |
Collapse
|
162
|
Jarmuszkiewicz W, Szewczyk A. Energy-dissipating hub in muscle mitochondria: Potassium channels and uncoupling proteins. Arch Biochem Biophys 2019; 664:102-109. [DOI: 10.1016/j.abb.2019.01.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 01/30/2019] [Accepted: 01/31/2019] [Indexed: 01/15/2023]
|
163
|
Nedergaard J, Cannon B. Brown adipose tissue as a heat-producing thermoeffector. HANDBOOK OF CLINICAL NEUROLOGY 2019; 156:137-152. [PMID: 30454587 DOI: 10.1016/b978-0-444-63912-7.00009-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Extra heat for defense of body temperature can be obtained from shivering or nonshivering thermogenesis. Nonshivering thermogenesis is a facultative (i.e., only occurring when needed) and adaptive (i.e., being augmented when the demand is chronically higher) process that, in mammals, is the result of the activity of uncoupling protein-1 (UCP1) in brown and brownish adipose tissues; no other quantitatively significant mechanism that fulfills the above criteria has been established. Measurement of heat production is generally indirect, based on oxygen consumption. Heat from brown adipose tissue is generated in mammals adapted to cold, in mammalian neonates, and in mammalian hibernators during arousal; brown adipose tissue may also be active in obese mammals and thus partially protect against further obesity. UCP1 is innately inhibited by cytosolic adenosine triphosphate (ATP) and is likely activated by fatty acids released from triglycerides within the cells; this lipolysis is stimulated by norepinephrine released from the sympathetic nerves innervating the tissue. For prolonged thermogenesis, substrate is delivered by the circulation as chylomicrons, lipoproteins, fatty acids, and glucose. The proton gradient over the mitochondrial membrane created by the respiratory chain is dispersed through the activity of UCP1; brown adipose tissue is nearly devoid of ATP synthase (as compared to respiratory chain activity). UCP1 developed likely at the dawn of mammalian evolution; most mammalian species still retain functional UCP1. Other members of the uncoupling protein family cannot uncouple. Both newborn and adult humans possess active brown adipose tissue but the significance of the tissue for adult human metabolism is not established.
Collapse
Affiliation(s)
- Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.
| | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
164
|
Ogunbona OB, Claypool SM. Emerging Roles in the Biogenesis of Cytochrome c Oxidase for Members of the Mitochondrial Carrier Family. Front Cell Dev Biol 2019; 7:3. [PMID: 30766870 PMCID: PMC6365663 DOI: 10.3389/fcell.2019.00003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/10/2019] [Indexed: 12/11/2022] Open
Abstract
The mitochondrial carrier family (MCF) is a group of transport proteins that are mostly localized to the inner mitochondrial membrane where they facilitate the movement of various solutes across the membrane. Although these carriers represent potential targets for therapeutic application and are repeatedly associated with human disease, research on the MCF has not progressed commensurate to their physiologic and pathophysiologic importance. Many of the 53 MCF members in humans are orphans and lack known transport substrates. Even for the relatively well-studied members of this family, such as the ADP/ATP carrier and the uncoupling protein, there exist fundamental gaps in our understanding of their biological roles including a clear rationale for the existence of multiple isoforms. Here, we briefly review this important family of mitochondrial carriers, provide a few salient examples of their diverse metabolic roles and disease associations, and then focus on an emerging link between several distinct MCF members, including the ADP/ATP carrier, and cytochrome c oxidase biogenesis. As the ADP/ATP carrier is regarded as the paradigm of the entire MCF, its newly established role in regulating translation of the mitochondrial genome highlights that we still have a lot to learn about these metabolite transporters.
Collapse
Affiliation(s)
- Oluwaseun B. Ogunbona
- Department of Physiology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
- Department of Pathology & Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Steven M. Claypool
- Department of Physiology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
165
|
Sreedhar A, Cassell T, Smith P, Lu D, Nam HW, Lane AN, Zhao Y. UCP2 Overexpression Redirects Glucose into Anabolic Metabolic Pathways. Proteomics 2019; 19:e1800353. [PMID: 30556651 DOI: 10.1002/pmic.201800353] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 11/19/2018] [Indexed: 01/05/2023]
Abstract
Uncoupling protein 2 (UCP2) is often upregulated in cancer cells. The UCP2 upregulation is positively correlated with enhanced proliferation, tumorigenesis, and metabolic alterations, thus suggesting that UCP2 upregulation can play a key role in sensing metabolic changes to promote tumorigenesis. To determine the global metabolic impact of UCP2 upregulation, 13 C6 glucose as a source molecule is used to "trace" the metabolic fate of carbon atoms derived from glucose. UCP2 overexpression in skin epidermal cells enhances the incorporation of 13 C label to pyruvate, tricarboxylic acid cycle intermediates, nucleotides, and amino acids, suggesting that UCP2 upregulation reprograms cellular metabolism toward macromolecule synthesis. To the best of our knowledge, this is the first study to bring to light the overall metabolic differences caused by UCP2 upregulation.
Collapse
Affiliation(s)
- Annapoorna Sreedhar
- Department of Pharmacology, Toxicology, and Neuroscience, LSU Health Sciences Center, Shreveport, LA, 71130, USA
| | - Teresa Cassell
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, 40509, USA
| | - Parker Smith
- Department of Pharmacology, Toxicology, and Neuroscience, LSU Health Sciences Center, Shreveport, LA, 71130, USA
| | - Daiwei Lu
- Department of Pharmacology, Toxicology, and Neuroscience, LSU Health Sciences Center, Shreveport, LA, 71130, USA
| | - Hyung W Nam
- Department of Pharmacology, Toxicology, and Neuroscience, LSU Health Sciences Center, Shreveport, LA, 71130, USA
| | - Andrew N Lane
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, KY, 40509, USA
| | - Yunfeng Zhao
- Department of Pharmacology, Toxicology, and Neuroscience, LSU Health Sciences Center, Shreveport, LA, 71130, USA
| |
Collapse
|
166
|
Molecular evolution of uncoupling proteins and implications for brain function. Neurosci Lett 2018; 696:140-145. [PMID: 30582970 DOI: 10.1016/j.neulet.2018.12.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 01/01/2023]
Abstract
Uncoupling proteins (UCPs) belong to the mitochondrial anion carrier superfamily and catalyze important metabolic functions at the mitochondrial inner membrane. While the thermogenic role of UCP1 in brown fat of eutherian mammals is well established, the molecular functions of UCP1 in ectothermic vertebrates and of other UCP paralogs remain less clear. Here, we critically discuss the existence of brain UCPs and their potential roles. Applying phylogenetic classification of novel UCPs, we summarize the evidence for brain UCP1 among vertebrates, the role of UCP2 in specific brain areas, and the existence of brain-specific UCPs. The phylogenetic analyses and discussion on functional data should alert the scientific community that the molecular function of so-called UCP1 homologues is by far not clarified and possibly relates to neither thermogenesis nor mitochondrial uncoupling.
Collapse
|
167
|
Mitochondrial Dynamics in Stem Cells and Differentiation. Int J Mol Sci 2018; 19:ijms19123893. [PMID: 30563106 PMCID: PMC6321186 DOI: 10.3390/ijms19123893] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 01/09/2023] Open
Abstract
Mitochondria are highly dynamic organelles that continuously change their shape. Their main function is adenosine triphosphate (ATP) production; however, they are additionally involved in a variety of cellular phenomena, such as apoptosis, cell cycle, proliferation, differentiation, reprogramming, and aging. The change in mitochondrial morphology is closely related to the functionality of mitochondria. Normal mitochondrial dynamics are critical for cellular function, embryonic development, and tissue formation. Thus, defects in proteins involved in mitochondrial dynamics that control mitochondrial fusion and fission can affect cellular differentiation, proliferation, cellular reprogramming, and aging. Here, we review the processes and proteins involved in mitochondrial dynamics and their various associated cellular phenomena.
Collapse
|
168
|
Abstract
Mitochondria are functionally versatile organelles. In addition to their conventional role of meeting the cell's energy requirements, mitochondria also actively regulate innate immune responses against infectious and sterile insults. Components of mitochondria, when released or exposed in response to dysfunction or damage, can be directly recognized by receptors of the innate immune system and trigger an immune response. In addition, despite initiation that may be independent from mitochondria, numerous innate immune responses are still subject to mitochondrial regulation as discrete steps of their signaling cascades occur on mitochondria or require mitochondrial components. Finally, mitochondrial metabolites and the metabolic state of the mitochondria within an innate immune cell modulate the precise immune response and shape the direction and character of that cell's response to stimuli. Together, these pathways result in a nuanced and very specific regulation of innate immune responses by mitochondria.
Collapse
Key Words
- ASC, Apoptosis Associated Speck like protein containing CARD
- ASK1, apoptosis signal-regulating kinase 1
- ATP, adenosine tri-phosphate
- CAPS, cryopyrin associated periodic syndromes
- CARD, caspase activation and recruitment domain
- CL, cardiolipin
- CLR, C-type lectin receptor
- CREB, cAMP response element binding protein
- Cgas, cyclic GMP-AMP synthase
- DAMP, damage associated molecular pattern
- ESCIT, evolutionarily conserved signaling intermediate in the toll pathway
- ETC, electron transport chain
- FPR, formyl peptide receptor
- HIF, hypoxia-inducible factor
- HMGB1, high mobility group box protein 1
- IFN, interferon
- IL, interleukin
- IRF, interferon regulatory factor
- JNK, cJUN NH2-terminal kinase
- LPS, lipopolysaccharide
- LRR, leucine rich repeat
- MAPK, mitogen-activated protein kinase
- MARCH5, membrane-associated ring finger (C3HC4) 5
- MAVS, mitochondrial antiviral signaling
- MAVS, mitochondrial antiviral signaling protein
- MFN1/2, mitofusin
- MOMP, mitochondrial outer membrane permeabilization
- MPT, mitochondrial permeability transition
- MyD88, myeloid differentiation primary response 88
- NADH, nicotinamide adenine dinucleotide
- NBD, nucleotide binding domain
- NFκB, Nuclear factor κ B
- NLR, NOD like receptor
- NOD, nucleotide-binding oligomerization domain
- NRF2, nuclear factor erythroid 2-related factor 2
- PAMP, pathogen associated molecular pattern
- PPAR, peroxisome proliferator-accelerated receptor
- PRRs, pathogen recognition receptors
- RIG-I, retinoic acid inducible gene I
- RLR, retinoic acid inducible gene like receptor
- ROS, reactive oxygen species
- STING, stimulator of interferon gene
- TAK1, transforming growth factor-β-activated kinase 1
- TANK, TRAF family member-associated NFκB activator
- TBK1, TANK Binding Kinase 1
- TCA, Tri-carboxylic acid
- TFAM, mitochondrial transcription factor A
- TLR, Toll Like Receptor
- TRAF6, tumor necrosis factor receptor-associated factor 6
- TRIF, TIR-domain-containing adapter-inducing interferon β
- TUFM, Tu translation elongation factor.
- fMet, N-formylated methionine
- mROS, mitochondrial ROS
- mtDNA, mitochondrial DNA
- n-fp, n-formyl peptides
Collapse
|
169
|
Banoth B, Cassel SL. Mitochondria in innate immune signaling. Transl Res 2018; 202:52-68. [PMID: 30165038 DOI: 10.1016/j.trsl.2018.07.014.mitochondria] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 05/25/2023]
Abstract
Mitochondria are functionally versatile organelles. In addition to their conventional role of meeting the cell's energy requirements, mitochondria also actively regulate innate immune responses against infectious and sterile insults. Components of mitochondria, when released or exposed in response to dysfunction or damage, can be directly recognized by receptors of the innate immune system and trigger an immune response. In addition, despite initiation that may be independent from mitochondria, numerous innate immune responses are still subject to mitochondrial regulation as discrete steps of their signaling cascades occur on mitochondria or require mitochondrial components. Finally, mitochondrial metabolites and the metabolic state of the mitochondria within an innate immune cell modulate the precise immune response and shape the direction and character of that cell's response to stimuli. Together, these pathways result in a nuanced and very specific regulation of innate immune responses by mitochondria.
Collapse
Key Words
- ASC, Apoptosis Associated Speck like protein containing CARD
- ASK1, apoptosis signal-regulating kinase 1
- ATP, adenosine tri-phosphate
- CAPS, cryopyrin associated periodic syndromes
- CARD, caspase activation and recruitment domain
- CL, cardiolipin
- CLR, C-type lectin receptor
- CREB, cAMP response element binding protein
- Cgas, cyclic GMP-AMP synthase
- DAMP, damage associated molecular pattern
- ESCIT, evolutionarily conserved signaling intermediate in the toll pathway
- ETC, electron transport chain
- FPR, formyl peptide receptor
- HIF, hypoxia-inducible factor
- HMGB1, high mobility group box protein 1
- IFN, interferon
- IL, interleukin
- IRF, interferon regulatory factor
- JNK, cJUN NH2-terminal kinase
- LPS, lipopolysaccharide
- LRR, leucine rich repeat
- MAPK, mitogen-activated protein kinase
- MARCH5, membrane-associated ring finger (C3HC4) 5
- MAVS, mitochondrial antiviral signaling
- MAVS, mitochondrial antiviral signaling protein
- MFN1/2, mitofusin
- MOMP, mitochondrial outer membrane permeabilization
- MPT, mitochondrial permeability transition
- MyD88, myeloid differentiation primary response 88
- NADH, nicotinamide adenine dinucleotide
- NBD, nucleotide binding domain
- NFκB, Nuclear factor κ B
- NLR, NOD like receptor
- NOD, nucleotide-binding oligomerization domain
- NRF2, nuclear factor erythroid 2-related factor 2
- PAMP, pathogen associated molecular pattern
- PPAR, peroxisome proliferator-accelerated receptor
- PRRs, pathogen recognition receptors
- RIG-I, retinoic acid inducible gene I
- RLR, retinoic acid inducible gene like receptor
- ROS, reactive oxygen species
- STING, stimulator of interferon gene
- TAK1, transforming growth factor-β-activated kinase 1
- TANK, TRAF family member-associated NFκB activator
- TBK1, TANK Binding Kinase 1
- TCA, Tri-carboxylic acid
- TFAM, mitochondrial transcription factor A
- TLR, Toll Like Receptor
- TRAF6, tumor necrosis factor receptor-associated factor 6
- TRIF, TIR-domain-containing adapter-inducing interferon β
- TUFM, Tu translation elongation factor.
- fMet, N-formylated methionine
- mROS, mitochondrial ROS
- mtDNA, mitochondrial DNA
- n-fp, n-formyl peptides
Collapse
Affiliation(s)
- Balaji Banoth
- Women's Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Suzanne L Cassel
- Women's Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California.
| |
Collapse
|
170
|
β3-Adrenoreceptors Control Mitochondrial Dormancy in Melanoma and Embryonic Stem Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6816508. [PMID: 30538804 PMCID: PMC6258109 DOI: 10.1155/2018/6816508] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/20/2018] [Indexed: 01/10/2023]
Abstract
The early phases of embryonic development and cancer share similar strategies to improve their survival in an inhospitable environment: both proliferate in a hypoxic and catecholamine-rich context, increasing aerobic glycolysis. Recent studies show that β3-adrenergic receptor (β3-AR) is involved in tumor progression, playing an important role in metastasis. Among β-adrenergic receptors, β3-AR is the last identified member of this family, and it is involved in cancer cell survival and induction of stromal reactivity in the tumor microenvironment. β3-AR is well known as a strong activator of uncoupling protein 1 (UCP1) in brown fat tissue. Interestingly, β3-AR is strongly expressed in early embryo development and in many cancer tissues. Induction of uncoupling protein 2 (UCP2) has been related to cancer metabolic switch, leading to accelerated glycolysis and reduced mitochondrial activity. In this study, for the first time, we demonstrate that β3-AR is able to promote this metabolic shift in both cancer and embryonic stem cells, inducing specific glycolytic cytoplasmic enzymes and a sort of mitochondrial dormancy through the induction of UCP2. The β3-AR/UCP2 axis induces a strong reduction of mitochondrial activity by reducing ATP synthesis and mitochondrial reactive oxygen species (mtROS) content. These effects are reverted by SR59230A, the specific β3-AR antagonist, causing an increase in mtROS. The increased level of mtROS is neutralized by a strong antioxidant activity in embryonic stem cells, but not in cancer stem cells, where it causes a dramatic reduction in tumor cell viability. These results lead to the possibility of a selective antitumor therapeutic use of SR59230A. Notably, we demonstrate the presence of β3-AR within the mitochondrial membrane in both cell lines, leading to the control of mitochondrial dormancy.
Collapse
|
171
|
Alkan HF, Walter KE, Luengo A, Madreiter-Sokolowski CT, Stryeck S, Lau AN, Al-Zoughbi W, Lewis CA, Thomas CJ, Hoefler G, Graier WF, Madl T, Vander Heiden MG, Bogner-Strauss JG. Cytosolic Aspartate Availability Determines Cell Survival When Glutamine Is Limiting. Cell Metab 2018; 28:706-720.e6. [PMID: 30122555 PMCID: PMC6390946 DOI: 10.1016/j.cmet.2018.07.021] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 05/29/2018] [Accepted: 07/29/2018] [Indexed: 12/23/2022]
Abstract
Mitochondrial function is important for aspartate biosynthesis in proliferating cells. Here, we show that mitochondrial aspartate export via the aspartate-glutamate carrier 1 (AGC1) supports cell proliferation and cellular redox homeostasis. Insufficient cytosolic aspartate delivery leads to cell death when TCA cycle carbon is reduced following glutamine withdrawal and/or glutaminase inhibition. Moreover, loss of AGC1 reduces allograft tumor growth that is further compromised by treatment with the glutaminase inhibitor CB-839. Together, these findings argue that mitochondrial aspartate export sustains cell survival in low-glutamine environments and AGC1 inhibition can synergize with glutaminase inhibition to limit tumor growth.
Collapse
Affiliation(s)
- H Furkan Alkan
- Institute of Biochemistry, Graz University of Technology, Humboldtstrasse 46/III, 8010 Graz, Austria; The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Katharina E Walter
- Institute of Biochemistry, Graz University of Technology, Humboldtstrasse 46/III, 8010 Graz, Austria
| | - Alba Luengo
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Corina T Madreiter-Sokolowski
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, A-8010 Graz, Austria
| | - Sarah Stryeck
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, A-8010 Graz, Austria
| | - Allison N Lau
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Wael Al-Zoughbi
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Neue Stiftingtalstraße 6, A-8010 Graz, Austria
| | - Caroline A Lewis
- Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA 02142, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA; Lymphoid Malignancies Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - Gerald Hoefler
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Neue Stiftingtalstraße 6, A-8010 Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Wolfgang F Graier
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, A-8010 Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Tobias Madl
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/6, A-8010 Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Matthew G Vander Heiden
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| | - Juliane G Bogner-Strauss
- Institute of Biochemistry, Graz University of Technology, Humboldtstrasse 46/III, 8010 Graz, Austria; BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
172
|
Cordani M, Butera G, Dando I, Torrens-Mas M, Butturini E, Pacchiana R, Oppici E, Cavallini C, Gasperini S, Tamassia N, Nadal-Serrano M, Coan M, Rossi D, Gaidano G, Caraglia M, Mariotto S, Spizzo R, Roca P, Oliver J, Scupoli MT, Donadelli M. Mutant p53 blocks SESN1/AMPK/PGC-1α/UCP2 axis increasing mitochondrial O 2-· production in cancer cells. Br J Cancer 2018; 119:994-1008. [PMID: 30318520 PMCID: PMC6203762 DOI: 10.1038/s41416-018-0288-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 09/11/2018] [Accepted: 09/14/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The TP53 tumor suppressor gene is the most frequently altered gene in tumors and mutant p53 gain-of-function isoforms actively promote cancer malignancy. METHODS A panel of wild-type and mutant p53 cancer cell lines of different tissues, including pancreas, breast, skin, and lung were used, as well as chronic lymphocytic leukemia (CLL) patients with different TP53 gene status. The effects of mutant p53 were evaluated by confocal microscopy, reactive oxygen species production assay, immunoblotting, and quantitative reverse transcription polymerase chain reaction after cellular transfection. RESULTS We demonstrate that oncogenic mutant p53 isoforms are able to inhibit SESN1 expression and consequently the amount of SESN1/AMPK complex, resulting in the downregulation of the AMPK/PGC-1α/UCP2 axis and mitochondrial O2-· production. We also show a correlation between the decrease of reduced thiols with a poorer clinical outcome of CLL patients bearing mutant TP53 gene. The restoration of the mitochondrial uncoupling protein 2 (UCP2) expression, as well as the addition of the radical scavenger N-acetyl-L-cysteine, reversed the oncogenic effects of mutant p53 as cellular hyper-proliferation, antiapoptotic effect, and resistance to drugs. CONCLUSIONS The inhibition of the SESN1/AMPK/PGC-1α/UCP2 axis contributes to the pro-oxidant and oncogenic effects of mutant p53, suggesting pro-oxidant drugs as a therapeutic approach for cancer patients bearing mutant TP53 gene.
Collapse
Affiliation(s)
- Marco Cordani
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy.,Biochemistry Department, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPAZ, Madrid, Spain
| | - Giovanna Butera
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Ilaria Dando
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Margalida Torrens-Mas
- Grupo Multidisciplinar de Oncología Traslacional, Instituto Universitario de Investigación en Ciencias de la Salud (IUNICS), Palma de Mallorca, Illes Balears, Spain.,Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria de Palma (IdISPa), Hospital Universitario Son Espases, edificio S. E-07120, Palma de Mallorca, Illes Balears, Spain
| | - Elena Butturini
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Raffaella Pacchiana
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Elisa Oppici
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Chiara Cavallini
- Research Center LURM (Interdepartmental Laboratory of Medical Research), University of Verona, Verona, Italy
| | - Sara Gasperini
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Nicola Tamassia
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | | | - Michela Coan
- Division of Molecular Oncology, Department of Translational Research, CRO National Cancer Institute Aviano, Aviano, Italy
| | - Davide Rossi
- Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland.,Institute of Oncology Research, Bellinzona, Switzerland
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Sofia Mariotto
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy
| | - Riccardo Spizzo
- Division of Molecular Oncology, Department of Translational Research, CRO National Cancer Institute Aviano, Aviano, Italy
| | - Pilar Roca
- Grupo Multidisciplinar de Oncología Traslacional, Instituto Universitario de Investigación en Ciencias de la Salud (IUNICS), Palma de Mallorca, Illes Balears, Spain.,Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria de Palma (IdISPa), Hospital Universitario Son Espases, edificio S. E-07120, Palma de Mallorca, Illes Balears, Spain
| | - Jordi Oliver
- Grupo Multidisciplinar de Oncología Traslacional, Instituto Universitario de Investigación en Ciencias de la Salud (IUNICS), Palma de Mallorca, Illes Balears, Spain.,Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria de Palma (IdISPa), Hospital Universitario Son Espases, edificio S. E-07120, Palma de Mallorca, Illes Balears, Spain
| | - Maria Teresa Scupoli
- Research Center LURM (Interdepartmental Laboratory of Medical Research), University of Verona, Verona, Italy
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Verona, Italy.
| |
Collapse
|
173
|
Jin S, Diano S. Mitochondrial Dynamics and Hypothalamic Regulation of Metabolism. Endocrinology 2018; 159:3596-3604. [PMID: 30203064 PMCID: PMC6157417 DOI: 10.1210/en.2018-00667] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/02/2018] [Indexed: 01/22/2023]
Abstract
Mitochondria are cellular organelles that play an important role in bioenergetic processes. In the central nervous system, high energy-demanding neurons are critically dependent on mitochondria to fulfill their appropriate functions. The hypothalamus is a key brain area for maintaining glucose and energy homeostasis via the ability of hypothalamic neurons to sense, integrate, and respond to numerous metabolic signals. Mitochondrial function has emerged as an important component in the regulation of hypothalamic neurons controlling glucose and energy homeostasis. Although the underlying mechanisms are not fully understood, emerging evidence indicates that mitochondrial dysfunction in hypothalamic neurons may contribute to the development of various metabolic diseases, including obesity and type 2 diabetes mellitus (T2DM). In this review, we summarize recent studies demonstrating the link between mitochondria and hypothalamic neural control of glucose and energy homeostasis. Finally, this review provides an insight to understand how mitochondria in hypothalamic neurons may contribute to the development of metabolic disorders, such as T2DM and obesity.
Collapse
Affiliation(s)
- Sungho Jin
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Sabrina Diano
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut
- Department of Clinical Medicine and Surgery, University of Naples “Federico II,” Naples, Italy
| |
Collapse
|
174
|
Zheng H, Yu WM, Shen J, Kang S, Hambardzumyan D, Li JY, Shen Y, Kenney AM, Chen J, Qu CK. Mitochondrial oxidation of the carbohydrate fuel is required for neural precursor/stem cell function and postnatal cerebellar development. SCIENCE ADVANCES 2018; 4:eaat2681. [PMID: 30338292 PMCID: PMC6191298 DOI: 10.1126/sciadv.aat2681] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 09/05/2018] [Indexed: 05/28/2023]
Abstract
While deregulation of mitochondrial metabolism and cytosolic glycolysis has been well recognized in tumor cells, the role of coordinated mitochondrial oxidation and cytosolic fermentation of pyruvate, a key metabolite derived from glucose, in physiological processes is not well understood. Here, we report that knockout of PTPMT1, a mitochondrial phosphoinositide phosphatase, completely blocked postnatal cerebellar development. Proliferation of granule cell progenitors, the most actively replicating cells in the developing cerebellum, was only moderately decreased, and proliferation of Purkinje cell progenitors did not seem to be affected in knockout mice. In contrast, generation of functional Bergmann glia from multipotent precursor cells (radial glia), which is essential for cerebellar corticogenesis, was totally disrupted. Moreover, despite a low turnover rate, neural stem cells were impaired in self-renewal in knockout mice. Mechanistically, loss of PTPMT1 decreased mitochondrial aerobic metabolism by limiting utilization of pyruvate, which resulted in bioenergetic stress in neural precursor/stem cells but not in progenitor or mature cells, leading to cell cycle arrest through activation of the AMPK-p19/p21 pathway. This study suggests that mitochondrial oxidation of the carbohydrate fuel is required for postnatal cerebellar development, and identifies a bioenergetic stress-induced cell cycle checkpoint in neural precursor/stem cells.
Collapse
Affiliation(s)
- Hong Zheng
- Department of Pediatrics, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA 30322, USA
- Department of Medicine, Center for Stem Cell and Regenerative Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Wen-Mei Yu
- Department of Pediatrics, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA 30322, USA
- Department of Medicine, Center for Stem Cell and Regenerative Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jinhua Shen
- Department of Medicine, Center for Stem Cell and Regenerative Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Sumin Kang
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA
| | - Dolores Hambardzumyan
- Department of Pediatrics, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA 30322, USA
| | - James Y. Li
- Department of Genetics and Genome Sciences, University of Connecticut, Farmington, CT 06030, USA
| | - Yuxian Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Anna M. Kenney
- Department of Pediatrics, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA 30322, USA
| | - Jing Chen
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA 30322, USA
| | - Cheng-Kui Qu
- Department of Pediatrics, Children’s Healthcare of Atlanta, Emory University, Atlanta, GA 30322, USA
- Department of Medicine, Center for Stem Cell and Regenerative Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
175
|
Lu HY, Zeng H, Zhang L, Porres JM, Cheng WH. Fecal fermentation products of common bean-derived fiber inhibit C/EBPα and PPARγ expression and lipid accumulation but stimulate PPARδ and UCP2 expression in the adipogenesis of 3T3-L1 cells. J Nutr Biochem 2018; 60:9-15. [DOI: 10.1016/j.jnutbio.2018.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 05/22/2018] [Accepted: 06/06/2018] [Indexed: 12/19/2022]
|
176
|
Malik D, Kaul D. Human cellular mitochondrial remodelling is governed by miR-2909 RNomics. PLoS One 2018; 13:e0203614. [PMID: 30252847 PMCID: PMC6155498 DOI: 10.1371/journal.pone.0203614] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 08/23/2018] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND There exists a general recognition of the fact that mitochondrial remodelling as a result of aerobic glycolysis ensures human somatic cells to revert to a more primitive-form exhibiting stem-like phenotype. The present study is an attempt to demonstrate that miR-2909 RNomics within human peripheral blood mononuclear cells (PBMCs) has the inherent capacity to re-program these cells to exhibit mitochondrial remodelling paralleled by aerobic glycolysis together with intracellular lipid inclusions. Such re-programmed PBMCs also expressed genes having ability to sustain their de-differentiation state and survival. MATERIAL AND METHODS Human PBMCs were programed to ectopically express miR-2909. Expression levels of genes including glucose transporter-1 (Glut-1), hexokinase (HK), hypoxia inducia factor-1 (HIF-1α), c-Myc, p53,mechanistic target of rapamycin complex (mTORC1), polycombcomplex protein (Bmi-1), Notch,Nanog,Tie-2, Oct-4,CD59, p53, CD34, B-cell lymphoma-2 (Bcl2),sterol regulatory element-binding protein2 (SREBP2), peroxisome proliferator-activated receptor gamma (PPARγ) nuclear respiratory factor 1 (NRF1), mitochondrial transcription factor A (Tfam), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) within miR-2909 expression vector transfected human PBMCs as well as PBMCs transfected with control vector containing scrambled sequence after 48h post-transfection using RT-qPCR and cellular ultrastructural features induced by miR-2909 ectopic expression were observed using transmission electron microscopy and morphometric analysis of an electron micrograph. RESULTS Ectopic expression of miR-2909 within human PBMCs resulted in their reprogramming into stem-like phenotype indicated by mitochondrial globular shaped coupled with cristae-poor morphology. Nuclear to cytoplasmic ratio (N/C), quantification of ATP levels, GSSG/GSH ratio, mitochondrial cytochrome c oxidase activity, secreted lactate concentrations, activity of antioxidant enzymes, levels of esterified cholesterol and triglycerides and flow-cytometric detection of apoptosis confirmed the compromised nature of mitochondrial function induced by ectopic miR-2909 expression in human PBMCs. CONCLUSION Based upon these results we propose that AATF gene-encoded miR-2909 may act as an epigenetic switch for cellular aerobic-glycolysis to ensure de-differentiation.
Collapse
Affiliation(s)
- Deepti Malik
- Molecular Biology Unit, Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh (India)
| | - Deepak Kaul
- Molecular Biology Unit, Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh (India)
| |
Collapse
|
177
|
Uncoupling proteins as a therapeutic target to protect the diabetic heart. Pharmacol Res 2018; 137:11-24. [PMID: 30223086 DOI: 10.1016/j.phrs.2018.09.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/07/2018] [Accepted: 09/13/2018] [Indexed: 12/16/2022]
Abstract
Myocardial remodeling and dysfunction caused by accelerated oxidative damage is a widely reported phenomenon within a diabetic state. Altered myocardial substrate preference appears to be the major cause of enhanced oxidative stress-mediated cell injury within a diabetic heart. During this process, exacerbated free fatty acid flux causes an abnormal increase in mitochondrial membrane potential leading to the overproduction of free radical species and subsequent cell damage. Uncoupling proteins (UCPs) are expressed within the myocardium and can protect against free radical damage by modulating mitochondrial respiration, leading to reduced production of reactive oxygen species. Moreover, transgenic animals lacking UCPs have been shown to be more susceptible to oxidative damage and display reduced cardiac function when compared to wild type animals. This suggests that tight regulation of UCPs is necessary for normal cardiac function and in the prevention of diabetes-induced oxidative damage. This review aims to enhance our understanding of the pathophysiological mechanisms relating to the role of UCPs in a diabetic heart, and further discuss known pharmacological compounds and hormones that can protect a diabetic heart through the modulation of UCPs.
Collapse
|
178
|
Ježek P, Holendová B, Garlid KD, Jabůrek M. Mitochondrial Uncoupling Proteins: Subtle Regulators of Cellular Redox Signaling. Antioxid Redox Signal 2018; 29:667-714. [PMID: 29351723 PMCID: PMC6071544 DOI: 10.1089/ars.2017.7225] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Mitochondria are the energetic, metabolic, redox, and information signaling centers of the cell. Substrate pressure, mitochondrial network dynamics, and cristae morphology state are integrated by the protonmotive force Δp or its potential component, ΔΨ, which are attenuated by proton backflux into the matrix, termed uncoupling. The mitochondrial uncoupling proteins (UCP1-5) play an eminent role in the regulation of each of the mentioned aspects, being involved in numerous physiological events including redox signaling. Recent Advances: UCP2 structure, including purine nucleotide and fatty acid (FA) binding sites, strongly support the FA cycling mechanism: UCP2 expels FA anions, whereas uncoupling is achieved by the membrane backflux of protonated FA. Nascent FAs, cleaved by phospholipases, are preferential. The resulting Δp dissipation decreases superoxide formation dependent on Δp. UCP-mediated antioxidant protection and its impairment are expected to play a major role in cell physiology and pathology. Moreover, UCP2-mediated aspartate, oxaloacetate, and malate antiport with phosphate is expected to alter metabolism of cancer cells. CRITICAL ISSUES A wide range of UCP antioxidant effects and participations in redox signaling have been reported; however, mechanisms of UCP activation are still debated. Switching off/on the UCP2 protonophoretic function might serve as redox signaling either by employing/releasing the extra capacity of cell antioxidant systems or by directly increasing/decreasing mitochondrial superoxide sources. Rapid UCP2 degradation, FA levels, elevation of purine nucleotides, decreased Mg2+, or increased pyruvate accumulation may initiate UCP-mediated redox signaling. FUTURE DIRECTIONS Issues such as UCP2 participation in glucose sensing, neuronal (synaptic) function, and immune cell activation should be elucidated. Antioxid. Redox Signal. 29, 667-714.
Collapse
Affiliation(s)
- Petr Ježek
- 1 Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences , Prague, Czech Republic
| | - Blanka Holendová
- 1 Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences , Prague, Czech Republic
| | - Keith D Garlid
- 2 UCLA Cardiovascular Research Laboratory, David Geffen School of Medicine at UCLA , Los Angeles, California
| | - Martin Jabůrek
- 1 Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences , Prague, Czech Republic
| |
Collapse
|
179
|
Cadenas S. Mitochondrial uncoupling, ROS generation and cardioprotection. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2018; 1859:940-950. [DOI: 10.1016/j.bbabio.2018.05.019] [Citation(s) in RCA: 238] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/11/2018] [Accepted: 05/29/2018] [Indexed: 12/31/2022]
|
180
|
Kandasamy P, Gyimesi G, Kanai Y, Hediger MA. Amino acid transporters revisited: New views in health and disease. Trends Biochem Sci 2018; 43:752-789. [PMID: 30177408 DOI: 10.1016/j.tibs.2018.05.003] [Citation(s) in RCA: 266] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 05/23/2018] [Accepted: 05/25/2018] [Indexed: 02/09/2023]
Abstract
Amino acid transporters (AATs) are membrane-bound transport proteins that mediate transfer of amino acids into and out of cells or cellular organelles. AATs have diverse functional roles ranging from neurotransmission to acid-base balance, intracellular energy metabolism, and anabolic and catabolic reactions. In cancer cells and diabetes, dysregulation of AATs leads to metabolic reprogramming, which changes intracellular amino acid levels, contributing to the pathogenesis of cancer, obesity and diabetes. Indeed, the neutral amino acid transporters (NATs) SLC7A5/LAT1 and SLC1A5/ASCT2 are likely involved in several human malignancies. However, a clinical therapy that directly targets AATs has not yet been developed. The purpose of this review is to highlight the structural and functional diversity of AATs, their diverse physiological roles in different tissues and organs, their wide-ranging implications in human diseases and the emerging strategies and tools that will be necessary to target AATs therapeutically.
Collapse
Affiliation(s)
- Palanivel Kandasamy
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, CH-3012 Bern, Switzerland
| | - Gergely Gyimesi
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, CH-3012 Bern, Switzerland
| | - Yoshikatsu Kanai
- Division of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Matthias A Hediger
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, CH-3012 Bern, Switzerland.
| |
Collapse
|
181
|
Porcelli V, Vozza A, Calcagnile V, Gorgoglione R, Arrigoni R, Fontanesi F, Marobbio CMT, Castegna A, Palmieri F, Palmieri L. Molecular identification and functional characterization of a novel glutamate transporter in yeast and plant mitochondria. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2018; 1859:1249-1258. [PMID: 30297026 DOI: 10.1016/j.bbabio.2018.08.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 11/19/2022]
Abstract
The genome of Saccharomyces cerevisiae encodes 35 members of the mitochondrial carrier family (MCF) and 58 MCF members are coded by the genome of Arabidopsis thaliana, most of which have been functionally characterized. Here two members of this family, Ymc2p from S. cerevisiae and BOU from Arabidopsis, have been thoroughly characterized. These proteins were overproduced in bacteria and reconstituted into liposomes. Their transport properties and kinetic parameters demonstrate that Ymc2p and BOU transport glutamate, and to a much lesser extent L-homocysteinesulfinate, but not other amino acids and many other tested metabolites. Transport catalyzed by both carriers was saturable, inhibited by mercuric chloride and dependent on the proton gradient across the proteoliposomal membrane. The growth phenotype of S. cerevisiae cells lacking the genes ymc2 and agc1, which encodes the only other S. cerevisiae carrier capable to transport glutamate besides aspartate, was fully complemented by expressing Ymc2p, Agc1p or BOU. Mitochondrial extracts derived from ymc2Δagc1Δ cells, reconstituted into liposomes, exhibited no glutamate transport at variance with wild-type, ymc2Δ and agc1Δ cells, showing that S. cerevisiae cells grown in the presence of acetate do not contain additional mitochondrial transporters for glutamate besides Ymc2p and Agc1p. Furthermore, mitochondria isolated from wild-type, ymc2Δ and agc1Δ strains, but not from the double mutant ymc2Δagc1Δ strain, swell in isosmotic ammonium glutamate showing that glutamate is transported by Ymc2p and Agc1p together with a H+. It is proposed that the function of Ymc2p and BOU is to transport glutamate across the mitochondrial inner membrane and thereby play a role in intermediary metabolism, C1 metabolism and mitochondrial protein synthesis.
Collapse
Affiliation(s)
- Vito Porcelli
- Laboratory of Biochemistry and Molecular Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Angelo Vozza
- Laboratory of Biochemistry and Molecular Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Valeria Calcagnile
- Laboratory of Biochemistry and Molecular Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Ruggiero Gorgoglione
- Laboratory of Biochemistry and Molecular Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Roberto Arrigoni
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Bari, Italy
| | - Flavia Fontanesi
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Carlo M T Marobbio
- Laboratory of Biochemistry and Molecular Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy; CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Bari, Italy
| | - Alessandra Castegna
- Laboratory of Biochemistry and Molecular Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy; CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Bari, Italy
| | - Ferdinando Palmieri
- Laboratory of Biochemistry and Molecular Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy; CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Bari, Italy
| | - Luigi Palmieri
- Laboratory of Biochemistry and Molecular Biology, Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy; CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), Bari, Italy.
| |
Collapse
|
182
|
Tian XY, Ma S, Tse G, Wong WT, Huang Y. Uncoupling Protein 2 in Cardiovascular Health and Disease. Front Physiol 2018; 9:1060. [PMID: 30116205 PMCID: PMC6082951 DOI: 10.3389/fphys.2018.01060] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 07/16/2018] [Indexed: 12/22/2022] Open
Abstract
Uncoupling protein 2 (UCP2) belongs to the family of mitochondrial anion carrier proteins. It uncouples oxygen consumption from ATP synthesis. UCP2 is ubiquitously expressed in most cell types to reduce oxidative stress. It is tightly regulated at the transcriptional, translational, and post-translational levels. UCP2 in the cardiovascular system is being increasingly recognized as an important molecule to defend against various stress signals such as oxidative stress in the pathology of vascular dysfunction, atherosclerosis, hypertension, and cardiac injuries. UCP2 protects against cellular dysfunction through reducing mitochondrial oxidative stress and modulation of mitochondrial function. In view of the different functions of UCP2 in various cell types that contribute to whole body homeostasis, cell type-specific modification of UCP2 expression may offer a better approach to help understanding how UCP2 governs mitochondrial function, reactive oxygen species production and transmembrane proton leak and how dysfunction of UCP2 participates in the development of cardiovascular diseases. This review article provided an update on the physiological regulation of UCP2 in the cardiovascular system, and also discussed the involvement of UCP2 deficiency and associated oxidative stress in the pathogenesis of several common cardiovascular diseases. Drugs targeting UCP2 expression and activity might serve another effective strategy to ameliorate cardiovascular dysfunction. However, more detailed mechanistic study will be needed to dissect the role of UCP2, the regulation of UCP2 expression, and the cellular responses to the changes of UCP2 expression in normal and stressed situations at different stages of cardiovascular diseases.
Collapse
Affiliation(s)
- Xiao Yu Tian
- School of Biomedical Sciences, Institute of Vascular Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Shuangtao Ma
- Division of Nanomedicine and Molecular Intervention, Department of Medicine, Michigan State University, East Lansing, MI, United States
| | - Gary Tse
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Wing Tak Wong
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu Huang
- School of Biomedical Sciences, Institute of Vascular Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
183
|
Corona-Herrera GA, Arranz SE, Martínez-Palacios CA, Navarrete-Ramírez P, Toledo-Cuevas EM, Valdez-Alarcón JJ, Martínez-Chávez CC. Experimental evidence of masculinization by continuous illumination in a temperature sex determination teleost (Atherinopsidae) model: is oxidative stress involved? JOURNAL OF FISH BIOLOGY 2018; 93:229-237. [PMID: 29931822 DOI: 10.1111/jfb.13651] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 05/14/2018] [Indexed: 06/08/2023]
Abstract
The present study evaluates the influence of continuous light on phenotypic sex ratios in Chirostoma estor, a temperature sex determination animal model. Relative gene expression levels of 5 day old larvae were performed on two early gonad differentiation genes (sox9 and foxl2), two stress axis activation genes (gcr1 and crf) and four reactive oxygen species (ROS) antagonist effector genes (sod2, ucp2, gsr and cat). Two light treatments were applied from fertilization; control (12L:12D) simulated natural photoperiod and a continuous illumination photoperiod. By the end of the trial (12 weeks after hatching), differentiated and normal gonads were clearly identifiable in both treatments by histological observations. Regarding sex ratio, 73% of phenotypic males were found in continuous illumination compared with 40% in controls. Consistently, the sox9 gene (involved in early testis differentiation) showed an over expression in 64% of the individual larvae analysed compared with foxl2 (ovarian differentiation) suggesting a masculinization tendency in continuous illumination. On the other hand, only 36% of individuals showed the same tendency in the control treatment consistent with phenotypic sex ratios found under normal culture conditions. Relative gene expression results did not show significant difference in sod2, ucp2 and gcr1 levels, but cat, gsr and crf showed significantly higher expression levels in the continuous illumination treatment suggesting that both, the stress axis and ROS response mechanisms were activated at this time. This study suggests, a link between continuous light, oxidative stress and environmental sex determination in vertebrates. However, further research is necessary to describe this possible upstream mechanism that may drive some aspects of sexual plasticity in vertebrates.
Collapse
Affiliation(s)
- Guillermo A Corona-Herrera
- Instituto de Investigaciones Agropecuarias y Forestales, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Silvia E Arranz
- Laboratorio de Biotecnología Acuática, Universidad Nacional de Rosario, Santa Fe, Argentina
| | - Carlos A Martínez-Palacios
- Instituto de Investigaciones Agropecuarias y Forestales, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Pamela Navarrete-Ramírez
- CONACyT-Instituto de Investigaciones Agropecuarias y Forestales, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Elva M Toledo-Cuevas
- Instituto de Investigaciones Agropecuarias y Forestales, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Juan J Valdez-Alarcón
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo. Km. 9.5 carretera Morelia-Zinapecuaro, Tarimbaro, Mexico
| | - Carlos C Martínez-Chávez
- Instituto de Investigaciones Agropecuarias y Forestales, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| |
Collapse
|
184
|
Bachmann M, Costa R, Peruzzo R, Prosdocimi E, Checchetto V, Leanza L. Targeting Mitochondrial Ion Channels to Fight Cancer. Int J Mol Sci 2018; 19:ijms19072060. [PMID: 30011966 PMCID: PMC6073807 DOI: 10.3390/ijms19072060] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 12/14/2022] Open
Abstract
In recent years, several experimental evidences have underlined a new role of ion channels in cancer development and progression. In particular, mitochondrial ion channels are arising as new oncological targets, since it has been proved that most of them show an altered expression during tumor development and the pharmacological targeting of some of them have been demonstrated to be able to modulate cancer growth and progression, both in vitro as well as in vivo in pre-clinical mouse models. In this scenario, pharmacology of mitochondrial ion channels would be in the near future a new frontier for the treatment of tumors. In this review, we discuss the new advances in the field, by focusing our attention on the improvements in new drug developments to target mitochondrial ion channels.
Collapse
Affiliation(s)
| | - Roberto Costa
- Department of Biology, University of Padova, 35131 Padova, Italy.
| | - Roberta Peruzzo
- Department of Biology, University of Padova, 35131 Padova, Italy.
| | - Elena Prosdocimi
- Department of Biology, University of Padova, 35131 Padova, Italy.
| | | | - Luigi Leanza
- Department of Biology, University of Padova, 35131 Padova, Italy.
| |
Collapse
|
185
|
Nacarelli T, Azar A, Altinok O, Orynbayeva Z, Sell C. Rapamycin increases oxidative metabolism and enhances metabolic flexibility in human cardiac fibroblasts. GeroScience 2018; 40:10.1007/s11357-018-0030-2. [PMID: 29931650 PMCID: PMC6060207 DOI: 10.1007/s11357-018-0030-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 06/14/2018] [Indexed: 01/07/2023] Open
Abstract
Inhibition of mTOR signaling using rapamycin has been shown to increase lifespan and healthspan in multiple model organisms; however, the precise mechanisms for the beneficial effects of rapamycin remain uncertain. We have previously reported that rapamycin delays senescence in human cells and that enhanced mitochondrial biogenesis and protection from mitochondrial stress is one component of the benefit provided by rapamycin treatment. Here, using two models of senescence, replicative senescence and senescence induced by the presence of the Hutchinson-Gilford progeria lamin A mutation, we report that senescence is accompanied by elevated glycolysis and increased oxidative phosphorylation, which are both reduced by rapamycin. Measurements of mitochondrial function indicate that direct mitochondria targets of rapamycin are succinate dehydrogenase and matrix alanine aminotransferase. Elevated activity of these enzymes could be part of complex mechanisms that enable mitochondria to resume their optimal oxidative phosphorylation and resist senescence. This interpretation is supported by the fact that rapamycin-treated cultures do not undergo a premature senescence in response to the replacement of glucose with galactose in the culture medium, which forces a greater reliance on oxidative phosphorylation. Additionally, long-term treatment with rapamycin increases expression of the mitochondrial carrier protein UCP2, which facilitates the movement of metabolic intermediates across the mitochondrial membrane. The results suggest that rapamycin impacts mitochondrial function both through direct interaction with the mitochondria and through altered gene expression of mitochondrial carrier proteins.
Collapse
Affiliation(s)
| | - Ashley Azar
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, 19102, USA
| | - Oya Altinok
- Department of Surgery, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, 19102, USA
| | - Zulfiya Orynbayeva
- Department of Surgery, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, 19102, USA
| | - Christian Sell
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
186
|
Beta-hydroxybutyrate (3-OHB) can influence the energetic phenotype of breast cancer cells, but does not impact their proliferation and the response to chemotherapy or radiation. Cancer Metab 2018; 6:8. [PMID: 29942509 PMCID: PMC5996481 DOI: 10.1186/s40170-018-0180-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 05/17/2018] [Indexed: 12/30/2022] Open
Abstract
Background Ketogenic diets (KDs) or short-term fasting are popular trends amongst supportive approaches for cancer patients. Beta-hydroxybutyrate (3-OHB) is the main physiological ketone body, whose concentration can reach plasma levels of 2–6 mM during KDs or fasting. The impact of 3-OHB on the biology of tumor cells described so far is contradictory. Therefore, we investigated the effect of a physiological concentration of 3 mM 3-OHB on metabolism, proliferation, and viability of breast cancer (BC) cells in vitro. Methods Seven different human BC cell lines (BT20, BT474, HBL100, MCF-7, MDA-MB 231, MDA-MB 468, and T47D) were cultured in medium with 5 mM glucose in the presence of 3 mM 3-OHB at mild hypoxia (5% oxygen) or normoxia (21% oxygen). Metabolic profiling was performed by quantification of the turnover of glucose, lactate, and 3-OHB and by Seahorse metabolic flux analysis. Expression of key enzymes of ketolysis as well as the main monocarboxylic acid transporter MCT2 and the glucose-transporter GLUT1 was analyzed by RT-qPCR and Western blotting. The effect of 3-OHB on short- and long-term cell proliferation as well as chemo- and radiosensitivity were also analyzed. Results 3-OHB significantly changed the oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) in BT20 cells resulting in a more oxidative energetic phenotype. MCF-7 and MDA-MB 468 cells had increased ECAR only in response to 3-OHB, while the other three cell types remained uninfluenced. All cells expressed MCT2 and GLUT1, thus being able to uptake the metabolites. The consumption of 3-OHB was not strongly linked to mRNA overexpression of key enzymes of ketolysis and did not correlate with lactate production and glucose consumption. Neither 3-OHB nor acetoacetate did interfere with proliferation. Further, 3-OHB incubation did not modify the response of the tested BC cell lines to chemotherapy or radiation. Conclusions We found that a physiological level of 3-OHB can change the energetic profile of some BC cell lines. However, 3-OHB failed to influence different biologic processes in these cells, e.g., cell proliferation and the response to common breast cancer chemotherapy and radiotherapy. Thus, we have no evidence that 3-OHB generally influences the biology of breast cancer cells in vitro. Electronic supplementary material The online version of this article (10.1186/s40170-018-0180-9) contains supplementary material, which is available to authorized users.
Collapse
|
187
|
Metabolic influence of walnut phenolic extract on mitochondria in a colon cancer stem cell model. Eur J Nutr 2018; 58:1635-1645. [PMID: 29740695 DOI: 10.1007/s00394-018-1708-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 05/01/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE Walnut phenolic extract (WPE) reduces proliferation and enhances differentiation of colon cancer stem cells (CSCs). The present study investigated the metabolic influence of WPE on the mitochondrial function of colon CSCs to determine its underlying mechanism. METHODS CD133+CD44+ HCT116 colon cancer cells were selected by fluorescence-activated cell sorting and were treated with or without 40 µg/mL WPE. RNA-sequencing (RNA-Seq) was performed to identify differentially expressed genes (DEGs), which were further validated with RT-PCR. WPE-induced alterations in mitochondrial function were investigated through a mitochondrial stress test by determining cellular oxygen consumption rate (OCR), an indicator of mitochondrial respiration, and extracellular acidification rate (ECAR), an indicator of glycolysis, which were further confirmed by glucose uptake and lactate production tests. RESULTS RNA-Seq analysis identified two major functional clusters: metabolic and mitochondrial clusters. WPE treatment shifted the metabolic profile of cells towards the glycolysis pathway (ΔECAR = 36.98 mpH/min/ptn, p = 0.02) and oxidative pathway (ΔOCR = 29.18 pmol/min/ptn, p = 0.00001). Serial mitochondrial stimulations using respiration modulators, oligomycin, carbonyl cyanide-4 (trifluoromethoxy) phenylhydrazone, and rotenone/antimycin A, found an increased potential of mitochondrial respiration (ΔOCR = 111.5 pmol/min/ptn, p = 0.0006). WPE treatment also increased glucose uptake (Δ = 0.39 pmol/µL, p = 0.002) and lactate production (Δ = 0.08 nmol/µL, p = 0.005). CONCLUSIONS WPE treatment shifts the mitochondrial metabolism of colon CSC towards more aerobic glycolysis, which might be associated with the alterations in the characteristics of colon CSC.
Collapse
|
188
|
Yang H, Sun R, Ma N, Liu Q, Sun X, Zi P, Wang J, Chao K, Yu L. Inhibition of nuclear factor-κB signal by pyrrolidine dithiocarbamate alleviates lipopolysaccharide-induced acute lung injury. Oncotarget 2018; 8:47296-47304. [PMID: 28521300 PMCID: PMC5564565 DOI: 10.18632/oncotarget.17624] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 04/11/2017] [Indexed: 12/20/2022] Open
Abstract
This study mainly studied the effect of inhibition of nuclear factor-κB (NF-κB) signal by pyrrolidine dithiocarbamate (PDTC) on lipopolysaccharide (LPS)-induced inflammatory response, oxidative stress, and mitochondrial dysfunction in a murine acute lung injury model. The results showed that LPS exposure activated NF-κB and its upstream proteins and caused lung inflammation, oxidative stress, and mitochondrial dysfunction in mice. While inhibition of NF-κB by PDTC adminstration alleviated LPS-induced generation of lymphocytes, IL-1β, and TNF-α. Malondialdehyde, a common oxidative product, was markedly reduced after PDTC treatment in LPS-challenged mice. Furthermore, PDTC alleviated LPS-induced mitochondrial dysfunction via improving ATP synthesis and uncoupling protein 2 expression. In conclusion, inhibition of NF-κB by PDTC alleviated LPS-induced acute lung injury via maintaining inflammatory status, oxidative balance, and mitochondrial function in mice.
Collapse
Affiliation(s)
- Hongfu Yang
- Critical Care Medical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Rongqing Sun
- Critical Care Medical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Ning Ma
- Critical Care Medical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Qilong Liu
- Critical Care Medical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Xiaoge Sun
- Critical Care Medical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Panpan Zi
- Critical Care Medical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Junsheng Wang
- Critical Care Medical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Ke Chao
- Critical Care Medical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| | - Lei Yu
- Critical Care Medical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, P.R. China
| |
Collapse
|
189
|
Lisowski P, Kannan P, Mlody B, Prigione A. Mitochondria and the dynamic control of stem cell homeostasis. EMBO Rep 2018; 19:embr.201745432. [PMID: 29661859 DOI: 10.15252/embr.201745432] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/22/2017] [Accepted: 03/21/2018] [Indexed: 12/12/2022] Open
Abstract
The maintenance of cellular identity requires continuous adaptation to environmental changes. This process is particularly critical for stem cells, which need to preserve their differentiation potential over time. Among the mechanisms responsible for regulating cellular homeostatic responses, mitochondria are emerging as key players. Given their dynamic and multifaceted role in energy metabolism, redox, and calcium balance, as well as cell death, mitochondria appear at the interface between environmental cues and the control of epigenetic identity. In this review, we describe how mitochondria have been implicated in the processes of acquisition and loss of stemness, with a specific focus on pluripotency. Dissecting the biological functions of mitochondria in stem cell homeostasis and differentiation will provide essential knowledge to understand the dynamics of cell fate modulation, and to establish improved stem cell-based medical applications.
Collapse
Affiliation(s)
- Pawel Lisowski
- Max Delbrueck Center for Molecular Medicine (MDC), Berlin, Germany.,Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Magdalenka, Poland.,Centre for Preclinical Research and Technology (CePT), Warsaw Medical University, Warsaw, Poland
| | - Preethi Kannan
- Max Delbrueck Center for Molecular Medicine (MDC), Berlin, Germany
| | - Barbara Mlody
- Max Delbrueck Center for Molecular Medicine (MDC), Berlin, Germany
| | | |
Collapse
|
190
|
Kapnick SM, Pacheco SE, McGuire PJ. The emerging role of immune dysfunction in mitochondrial diseases as a paradigm for understanding immunometabolism. Metabolism 2018; 81:97-112. [PMID: 29162500 PMCID: PMC5866745 DOI: 10.1016/j.metabol.2017.11.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 11/07/2017] [Accepted: 11/11/2017] [Indexed: 01/08/2023]
Abstract
Immunometabolism aims to define the role of intermediary metabolism in immune cell function, with bioenergetics and the mitochondria recently taking center stage. To date, the medical literature on mitochondria and immune function extols the virtues of mouse models in exploring this biologic intersection. While the laboratory mouse has become a standard for studying mammalian biology, this model comprises part of a comprehensive approach. Humans, with their broad array of inherited phenotypes, serve as a starting point for studying immunometabolism; specifically, patients with mitochondrial disease. Using this top-down approach, the mouse as a model organism facilitates further exploration of the consequences of mutations involved in mitochondrial maintenance and function. In this review, we will discuss the emerging phenotype of immune dysfunction in mitochondrial disease as a model for understanding the role of the mitochondria in immune function in available mouse models.
Collapse
Affiliation(s)
- Senta M Kapnick
- Metabolism, Infection and Immunity Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Susan E Pacheco
- Department of Pediatrics, The University of Texas Health Science Center, Houston, TX, USA
| | - Peter J McGuire
- Metabolism, Infection and Immunity Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
191
|
Szelechowski M, Amoedo N, Obre E, Léger C, Allard L, Bonneu M, Claverol S, Lacombe D, Oliet S, Chevallier S, Le Masson G, Rossignol R. Metabolic Reprogramming in Amyotrophic Lateral Sclerosis. Sci Rep 2018; 8:3953. [PMID: 29500423 PMCID: PMC5834494 DOI: 10.1038/s41598-018-22318-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 02/21/2018] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial dysfunction in the spinal cord is a hallmark of amyotrophic lateral sclerosis (ALS), but the neurometabolic alterations during early stages of the disease remain unknown. Here, we investigated the bioenergetic and proteomic changes in ALS mouse motor neurons and patients' skin fibroblasts. We first observed that SODG93A mice presymptomatic motor neurons display alterations in the coupling efficiency of oxidative phosphorylation, along with fragmentation of the mitochondrial network. The proteome of presymptomatic ALS mice motor neurons also revealed a peculiar metabolic signature with upregulation of most energy-transducing enzymes, including the fatty acid oxidation (FAO) and the ketogenic components HADHA and ACAT2, respectively. Accordingly, FAO inhibition altered cell viability specifically in ALS mice motor neurons, while uncoupling protein 2 (UCP2) inhibition recovered cellular ATP levels and mitochondrial network morphology. These findings suggest a novel hypothesis of ALS bioenergetics linking FAO and UCP2. Lastly, we provide a unique set of data comparing the molecular alterations found in human ALS patients' skin fibroblasts and SODG93A mouse motor neurons, revealing conserved changes in protein translation, folding and assembly, tRNA aminoacylation and cell adhesion processes.
Collapse
Affiliation(s)
- M Szelechowski
- INSERM U1215, Neurocentre Magendie, 33077, Bordeaux, cedex, France
- Bordeaux University, 33000, Bordeaux, France
| | - N Amoedo
- Bordeaux University, 33000, Bordeaux, France
- INSERM U1211, MRGM, 33000, Bordeaux, France
| | - E Obre
- CELLOMET, Center of Functional Genomics (CGFB), 146 Rue Léo Saignat, 33000, Bordeaux, France
| | - C Léger
- INSERM U1215, Neurocentre Magendie, 33077, Bordeaux, cedex, France
- Bordeaux University, 33000, Bordeaux, France
| | - L Allard
- INSERM U1215, Neurocentre Magendie, 33077, Bordeaux, cedex, France
- Bordeaux University, 33000, Bordeaux, France
| | - M Bonneu
- Bordeaux University, 33000, Bordeaux, France
- Center of Functional Genomics (CGFB), Proteomic Facility, Bordeaux University, 33000, Bordeaux, France
| | - S Claverol
- Bordeaux University, 33000, Bordeaux, France
- Center of Functional Genomics (CGFB), Proteomic Facility, Bordeaux University, 33000, Bordeaux, France
| | - D Lacombe
- Bordeaux University, 33000, Bordeaux, France
- INSERM U1211, MRGM, 33000, Bordeaux, France
| | - S Oliet
- INSERM U1215, Neurocentre Magendie, 33077, Bordeaux, cedex, France
- Bordeaux University, 33000, Bordeaux, France
| | - S Chevallier
- INSERM U1215, Neurocentre Magendie, 33077, Bordeaux, cedex, France
- Bordeaux University, 33000, Bordeaux, France
| | - G Le Masson
- INSERM U1215, Neurocentre Magendie, 33077, Bordeaux, cedex, France.
- Bordeaux University, 33000, Bordeaux, France.
| | - R Rossignol
- Bordeaux University, 33000, Bordeaux, France.
- INSERM U1211, MRGM, 33000, Bordeaux, France.
| |
Collapse
|
192
|
Dahan P, Lu V, Nguyen RMT, Kennedy SAL, Teitell MA. Metabolism in pluripotency: Both driver and passenger? J Biol Chem 2018; 294:5420-5429. [PMID: 29463682 DOI: 10.1074/jbc.tm117.000832] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Pluripotent stem cells (PSCs) are highly proliferative cells characterized by robust metabolic demands to power rapid division. For many years considered a passive component or "passenger" of cell-fate determination, cell metabolism is now starting to take center stage as a driver of cell fate outcomes. This review provides an update and analysis of our current understanding of PSC metabolism and its role in self-renewal, differentiation, and somatic cell reprogramming to pluripotency. Moreover, we present evidence on the active roles metabolism plays in shaping the epigenome to influence patterns of gene expression that may model key features of early embryonic development.
Collapse
Affiliation(s)
- Perrine Dahan
- From the Departments of Pathology and Laboratory Medicine and
| | - Vivian Lu
- Molecular and Medical Pharmacology and
| | | | - Stephanie A L Kennedy
- From the Departments of Pathology and Laboratory Medicine and.,the Department of Biology, California State University at Northridge, Northridge, California 91330
| | - Michael A Teitell
- From the Departments of Pathology and Laboratory Medicine and .,the California NanoSystems Institute.,Department of Bioengineering, and.,Molecular Biology Institute, UCLA, Los Angeles, California 90095, and.,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, UCLA, Los Angeles, California 90095.,the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, California 90095
| |
Collapse
|
193
|
Abstract
Brown fat is emerging as an interesting and promising target for therapeutic intervention in obesity and metabolic disease. Activation of brown fat in humans is associated with marked improvement in metabolic parameters such as levels of free fatty acids and insulin sensitivity. Skeletal muscle is another important organ for thermogenesis, with the capacity to induce energy-consuming futile cycles. In this Review, we focus on how these two major thermogenic organs - brown fat and muscle - act and cooperate to maintain normal body temperature. Moreover, in the light of disease-relevant mechanisms, we explore the molecular pathways that regulate thermogenesis in brown fat and muscle. Brown adipocytes possess a unique cellular mechanism to convert chemical energy into heat: uncoupling protein 1 (UCP1), which can short-circuit the mitochondrial proton gradient. However, recent research demonstrates the existence of several other energy-expending 'futile' cycles in both adipocytes and muscle, such as creatine and calcium cycling. These mechanisms can complement or even substitute for UCP1-mediated thermogenesis. Moreover, they expand our view of cold-induced thermogenesis from a special feature of brown adipocytes to a more general physiological principle. Finally, we discuss how thermogenic mechanisms can be exploited to expend energy and hence offer new therapeutic opportunities.
Collapse
Affiliation(s)
- Matthias J Betz
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Sven Enerbäck
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 9A, SE-405 30 Gothenburg, Sweden
| |
Collapse
|
194
|
Fouladiha H, Marashi SA, Shokrgozar MA, Farokhi M, Atashi A. Applications of a metabolic network model of mesenchymal stem cells for controlling cell proliferation and differentiation. Cytotechnology 2018; 70:331-338. [PMID: 28980092 PMCID: PMC5809662 DOI: 10.1007/s10616-017-0148-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 09/16/2017] [Indexed: 12/27/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can be isolated from several tissues of adults. In addition, MSCs have the potential of differentiation into several cell types. Therefore, MSCs are very useful in stem cell therapy and regenerative medicine. MSCs have also been used as gene or protein carriers. As a result, maintaining MSCs in a desirable metabolic state has been the subject of several studies. Here, we used a genome scale metabolic network model of bone marrow derived MSCs for exploring the metabolism of these cells. We analyzed metabolic fluxes of the model in order to find ways of increasing stem cell proliferation and differentiation. Consequently, the experimental results were in consistency with computational results. Therefore, analyzing metabolic models was proven to be a promising field in biomedical researches of stem cells.
Collapse
Affiliation(s)
- Hamideh Fouladiha
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Sayed-Amir Marashi
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran.
| | | | - Mehdi Farokhi
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Amir Atashi
- Stem Cell and Tissue Engineering Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
195
|
Monné M, Daddabbo L, Gagneul D, Obata T, Hielscher B, Palmieri L, Miniero DV, Fernie AR, Weber APM, Palmieri F. Uncoupling proteins 1 and 2 (UCP1 and UCP2) from Arabidopsis thaliana are mitochondrial transporters of aspartate, glutamate, and dicarboxylates. J Biol Chem 2018; 293:4213-4227. [PMID: 29371401 DOI: 10.1074/jbc.ra117.000771] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 01/15/2018] [Indexed: 12/29/2022] Open
Abstract
The Arabidopsis thaliana genome contains 58 members of the solute carrier family SLC25, also called the mitochondrial carrier family, many of which have been shown to transport specific metabolites, nucleotides, and cofactors across the mitochondrial membrane. Here, two Arabidopsis members of this family, AtUCP1 and AtUCP2, which were previously thought to be uncoupling proteins and hence named UCP1/PUMP1 and UCP2/PUMP2, respectively, are assigned with a novel function. They were expressed in bacteria, purified, and reconstituted in phospholipid vesicles. Their transport properties demonstrate that they transport amino acids (aspartate, glutamate, cysteine sulfinate, and cysteate), dicarboxylates (malate, oxaloacetate, and 2-oxoglutarate), phosphate, sulfate, and thiosulfate. Transport was saturable and inhibited by mercurials and other mitochondrial carrier inhibitors to various degrees. AtUCP1 and AtUCP2 catalyzed a fast counterexchange transport as well as a low uniport of substrates, with transport rates of AtUCP1 being much higher than those of AtUCP2 in both cases. The aspartate/glutamate heteroexchange mediated by AtUCP1 and AtUCP2 is electroneutral, in contrast to that mediated by the mammalian mitochondrial aspartate glutamate carrier. Furthermore, both carriers were found to be targeted to mitochondria. Metabolite profiling of single and double knockouts shows changes in organic acid and amino acid levels. Notably, AtUCP1 and AtUCP2 are the first reported mitochondrial carriers in Arabidopsis to transport aspartate and glutamate. It is proposed that the primary function of AtUCP1 and AtUCP2 is to catalyze an aspartateout/glutamatein exchange across the mitochondrial membrane and thereby contribute to the export of reducing equivalents from the mitochondria in photorespiration.
Collapse
Affiliation(s)
- Magnus Monné
- From the Department of Biosciences, Biotechnologies and Biopharmaceutics, Laboratory of Biochemistry and Molecular Biology, University of Bari, via Orabona 4, 70125 Bari, Italy.,the Department of Sciences, University of Basilicata, Via Ateneo Lucano 10, 85100 Potenza, Italy
| | - Lucia Daddabbo
- From the Department of Biosciences, Biotechnologies and Biopharmaceutics, Laboratory of Biochemistry and Molecular Biology, University of Bari, via Orabona 4, 70125 Bari, Italy
| | - David Gagneul
- the Cluster of Excellence on Plant Science (CEPLAS), Institute of Plant Biochemistry, Heinrich-Heine-Universität, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Toshihiro Obata
- the Department Willmitzer, Max-Planck-Institut fur Molekulare Pflanzenphysiologie, Am Muhlenberg 1, 14476 Potsdam-Golm, Germany, and
| | - Björn Hielscher
- the Cluster of Excellence on Plant Science (CEPLAS), Institute of Plant Biochemistry, Heinrich-Heine-Universität, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Luigi Palmieri
- From the Department of Biosciences, Biotechnologies and Biopharmaceutics, Laboratory of Biochemistry and Molecular Biology, University of Bari, via Orabona 4, 70125 Bari, Italy.,the Center of Excellence in Comparative Genomics, University of Bari, via Orabona 4, 70125 Bari, Italy
| | - Daniela Valeria Miniero
- From the Department of Biosciences, Biotechnologies and Biopharmaceutics, Laboratory of Biochemistry and Molecular Biology, University of Bari, via Orabona 4, 70125 Bari, Italy
| | - Alisdair R Fernie
- the Department Willmitzer, Max-Planck-Institut fur Molekulare Pflanzenphysiologie, Am Muhlenberg 1, 14476 Potsdam-Golm, Germany, and
| | - Andreas P M Weber
- the Cluster of Excellence on Plant Science (CEPLAS), Institute of Plant Biochemistry, Heinrich-Heine-Universität, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Ferdinando Palmieri
- From the Department of Biosciences, Biotechnologies and Biopharmaceutics, Laboratory of Biochemistry and Molecular Biology, University of Bari, via Orabona 4, 70125 Bari, Italy, .,the Center of Excellence in Comparative Genomics, University of Bari, via Orabona 4, 70125 Bari, Italy
| |
Collapse
|
196
|
Defects in the mitochondrial-tRNA modification enzymes MTO1 and GTPBP3 promote different metabolic reprogramming through a HIF-PPARγ-UCP2-AMPK axis. Sci Rep 2018; 8:1163. [PMID: 29348686 PMCID: PMC5773609 DOI: 10.1038/s41598-018-19587-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 01/04/2018] [Indexed: 12/12/2022] Open
Abstract
Human proteins MTO1 and GTPBP3 are thought to jointly catalyze the modification of the wobble uridine in mitochondrial tRNAs. Defects in each protein cause infantile hypertrophic cardiomyopathy with lactic acidosis. However, the underlying mechanisms are mostly unknown. Using fibroblasts from an MTO1 patient and MTO1 silenced cells, we found that the MTO1 deficiency is associated with a metabolic reprogramming mediated by inactivation of AMPK, down regulation of the uncoupling protein 2 (UCP2) and transcription factor PPARγ, and activation of the hypoxia inducible factor 1 (HIF-1). As a result, glycolysis and oxidative phosphorylation are uncoupled, while fatty acid metabolism is altered, leading to accumulation of lipid droplets in MTO1 fibroblasts. Unexpectedly, this response is different from that triggered by the GTPBP3 defect, as GTPBP3-depleted cells exhibit AMPK activation, increased levels of UCP2 and PPARγ, and inactivation of HIF-1. In addition, fatty acid oxidation and respiration are stimulated in these cells. Therefore, the HIF-PPARγ-UCP2-AMPK axis is operating differently in MTO1- and GTPBP3-defective cells, which strongly suggests that one of these proteins has an additional role, besides mitochondrial-tRNA modification. This work provides new and useful information on the molecular basis of the MTO1 and GTPBP3 defects and on putative targets for therapeutic intervention.
Collapse
|
197
|
Broche B, Ben Fradj S, Aguilar E, Sancerni T, Bénard M, Makaci F, Berthault C, Scharfmann R, Alves-Guerra MC, Duvillié B. Mitochondrial Protein UCP2 Controls Pancreas Development. Diabetes 2018; 67:78-84. [PMID: 29079704 DOI: 10.2337/db17-0118] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 10/23/2017] [Indexed: 11/13/2022]
Abstract
The mitochondrial carrier uncoupling protein (UCP) 2 belongs to the family of the UCPs. Despite its name, it is now accepted that UCP2 is rather a metabolite transporter than a UCP. UCP2 can regulate oxidative stress and/or energetic metabolism. In rodents, UCP2 is involved in the control of α- and β-cell mass as well as insulin and glucagon secretion. Our aim was to determine whether the effects of UCP2 observed on β-cell mass have an embryonic origin. Thus, we used Ucp2 knockout mice. We found an increased size of the pancreas in Ucp2-/- fetuses at embryonic day 16.5, associated with a higher number of α- and β-cells. This phenotype was caused by an increase of PDX1+ progenitor cells. Perinatally, an increase in the proliferation of endocrine cells also participates in their expansion. Next, we analyzed the oxidative stress in the pancreata. We quantified an increased nuclear translocation of nuclear factor erythroid 2-related factor 2 (NRF2) in the mutant, suggesting an increased production of reactive oxygen species (ROS). Phosphorylation of AKT, an ROS target, was also activated in the Ucp2-/- pancreata. Finally, administration of the antioxidant N-acetyl-l-cysteine to Ucp2-/- pregnant mice alleviated the effect of knocking out UCP2 on pancreas development. Together, these data demonstrate that UCP2 controls pancreas development through the ROS-AKT signaling pathway.
Collapse
Affiliation(s)
- Benjamin Broche
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Selma Ben Fradj
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Esther Aguilar
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Tiphaine Sancerni
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Matthieu Bénard
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Fatna Makaci
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Claire Berthault
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Raphaël Scharfmann
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marie-Clotilde Alves-Guerra
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Bertrand Duvillié
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
198
|
Hassan NE, El-Masry SA, Zarouk W, El Banna RA, Mosaad RM, Al-Tohamy M, Salamah AR. Obesity phenotype in relation to gene polymorphism among samples of Egyptian children and their mothers. Genes Dis 2017; 5:150-157. [PMID: 30258944 PMCID: PMC6146265 DOI: 10.1016/j.gendis.2017.12.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 12/06/2017] [Indexed: 02/06/2023] Open
Abstract
Obesity is complex heterogeneous disease controlled by genes, environmental factors, and their interaction. Genetic factors account for 40-90% of the body mass index variations. Body mass index (BMI) of children correlates more closely with maternal than paternal BMI. So, this studu was aimed to investigate the role of leptin receptor LEPR Gln223Arg, the uncoupling protein 2 (UCP2 G 866 A) and insulin receptor gene (INSR exon 17) polymorphisms in the pathogenesis of obesity. A cross-sectional study executed on 130 children and their obese mothers; classified into 2 groups according to their BMI. The 2 groups were evaluated regarding the anthropometry. Restriction fragment length analysis for LEPR Gln223Arg, UCP2 -866 G/A and INSR exon 17 polymorphisms were applied. It was reported that increased risk of obesity was found in LEPR AG + AA genotype and the A allele. Significant statistical difference was detected only in female children. Concerning UCP2, the AG followed by the GG genotype was the most frequent in all groups and the G allele was the mostly present in obese mothers and obese male children but with no statistical significance. There was difference in the INSR genotype and alleles between groups, but this difference was not statistically significant. This study concluded that the LEPR Gln223Arg, UCP2 G 866 A and INSR exon 17 polymorphisms are related to obesity in Egyptian population. Further researches on larger population are recommended to ascertain the implications of LEPR, UCP2 and INSR polymorphisms in obesity.
Collapse
Affiliation(s)
- Nayera E Hassan
- Biological Anthropology Department, Medical Research Division, National Research Centre, Giza, Egypt
| | - Sahar A El-Masry
- Biological Anthropology Department, Medical Research Division, National Research Centre, Giza, Egypt
| | - Waheba Zarouk
- Molecular Genetics and Enzymology Department, Human Genetics and Genomic Research Division, National Research Centre, Giza, Egypt
| | - Rokia A El Banna
- Biological Anthropology Department, Medical Research Division, National Research Centre, Giza, Egypt
| | - Rehab M Mosaad
- Molecular Genetics and Enzymology Department, Human Genetics and Genomic Research Division, National Research Centre, Giza, Egypt
| | - Muhammad Al-Tohamy
- Biological Anthropology Department, Medical Research Division, National Research Centre, Giza, Egypt
| | - Abeer Ramadan Salamah
- Molecular Genetics and Enzymology Department, Human Genetics and Genomic Research Division, National Research Centre, Giza, Egypt
| |
Collapse
|
199
|
Lu M, Li C. Nutrient sensing in pancreatic islets: lessons from congenital hyperinsulinism and monogenic diabetes. Ann N Y Acad Sci 2017; 1411:65-82. [PMID: 29044608 DOI: 10.1111/nyas.13448] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/05/2017] [Accepted: 07/14/2017] [Indexed: 12/14/2022]
Abstract
Pancreatic beta cells sense changes in nutrients during the cycles of fasting and feeding and release insulin accordingly to maintain glucose homeostasis. Abnormal beta cell nutrient sensing resulting from gene mutations leads to hypoglycemia or diabetes. Glucokinase (GCK) plays a key role in beta cell glucose sensing. As one form of congenital hyperinsulinism (CHI), activating mutations of GCK result in a decreased threshold for glucose-stimulated insulin secretion and hypoglycemia. In contrast, inactivating mutations of GCK result in diabetes, including a mild form (MODY2) and a severe form (permanent neonatal diabetes mellitus (PNDM)). Mutations of beta cell ion channels involved in insulin secretion regulation also alter glucose sensing. Activating or inactivating mutations of ATP-dependent potassium (KATP ) channel genes result in severe but completely opposite clinical phenotypes, including PNDM and CHI. Mutations of the other ion channels, including voltage-gated potassium channels (Kv 7.1) and voltage-gated calcium channels, also lead to abnormal glucose sensing and CHI. Furthermore, amino acids can stimulate insulin secretion in a glucose-independent manner in some forms of CHI, including activating mutations of the glutamate dehydrogenase gene, HDAH deficiency, and inactivating mutations of KATP channel genes. These genetic defects have provided insight into a better understanding of the complicated nature of beta cell fuel-sensing mechanisms.
Collapse
Affiliation(s)
- Ming Lu
- Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,Department of Pediatrics & Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China
| | - Changhong Li
- Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,Department of Pediatrics & Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
200
|
Abstract
PURPOSE Radiotherapy (RT) is a mainstay in the treatment of solid tumors and works by inducing free radical stress in tumor cells, leading to loss of reproductive integrity. The optimal treatment strategy has to consider damage to both tumor and normal cells and is determined by five factors known as the 5 R's of radiobiology: Reoxygenation, DNA repair, radiosensitivity, redistribution in the cell cycle and repopulation. The aim of this review is (i) to present evidence that these 5 R's are strongly influenced by cellular and whole-body metabolism that in turn can be modified through ketogenic therapy in form of ketogenic diets and short-term fasting and (ii) to stimulate new research into this field including some research questions deserving further study. CONCLUSIONS Preclinical and some preliminary clinical data support the hypothesis that ketogenic therapy could be utilized as a complementary treatment in order to improve the outcome after RT, both in terms of higher tumor control and in terms of lower normal tissue complication probability. The first effect relates to the metabolic shift from glycolysis toward mitochondrial metabolism that selectively increases ROS production and impairs ATP production in tumor cells. The second effect is based on the differential stress resistance phenomenon, which is achieved when glucose and growth factors are reduced and ketone bodies are elevated, reprogramming normal but not tumor cells from proliferation toward maintenance and stress resistance. Underlying both effects are metabolic differences between normal and tumor cells that ketogenic therapy seeks to exploit. Specifically, the recently discovered role of the ketone body β-hydroxybutyrate as an endogenous class-I histone deacetylase inhibitor suggests a dual role as a radioprotector of normal cells and a radiosensitzer of tumor cells that opens up exciting possibilities to employ ketogenic therapy as a cost-effective adjunct to radiotherapy against cancer.
Collapse
Affiliation(s)
- Rainer J Klement
- a Department of Radiotherapy and Radiation Oncology , Leopoldina Hospital , Schweinfurt , Germany
| |
Collapse
|