1
|
Attema B, de la Rosa Rodriguez MA, van Schothorst EM, Grefte S, Hooiveld GJ, Kersten S. Deficiency of the mitochondrial transporter SLC25A47 minimally impacts hepatic lipid metabolism in fasted and diet-induced obese mice. Mol Metab 2024; 92:102092. [PMID: 39746607 PMCID: PMC11773045 DOI: 10.1016/j.molmet.2024.102092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/20/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025] Open
Abstract
OBJECTIVE The peroxisome proliferator-activated receptor-alpha (PPARα) plays a central role in lipid metabolism in the liver by stimulating the expression of hundreds of genes. Accordingly, regulation by PPARα could be a screening tool to identify novel genes involved in hepatic lipid metabolism. Previously, the mitochondrial transporter SLC25A47 was suggested to play a role in energy metabolism and liver-specific uncoupling, but further research is lacking. METHODS We explored the potential role of SLC25A47 through in vitro studies and using mice overexpressing and lacking SLC25A47. RESULTS SLC25A47 was identified as a PPARα-regulated and fasting-induced gene in human and mouse hepatocytes. Adenoviral-mediated overexpression of SLC25A47 minimally impacted metabolic parameters during fasting and high-fat feeding. During high-fat feeding, SLC25A47 ablation also did not influence any metabolic parameters, apart from a minor improvement in glucose tolerance. In fasted mice, SLC25A47 ablation was associated with modest, reproducible, and likely indirect reductions in plasma triglycerides and glycerol. SLC25A47 ablation did not influence energy expenditure. Depending on the nutritional status, metabolomic analysis showed modest alterations in plasma, liver, and hepatic mitochondrial levels of various metabolites related to amino acid metabolism, TCA cycle, and fatty acid metabolism. No major and consistent alterations in levels of specific metabolites were found that establish the substrate for and function of SLC25A47. CONCLUSION Collectively, our results hint at a role of SLC25A47 in amino acid and fatty acid metabolism, yet suggest that SLC25A47 is dispensable for hepatic lipid homeostasis during fasting and high-fat feeding.
Collapse
Affiliation(s)
- Brecht Attema
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Montserrat A de la Rosa Rodriguez
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | | | - Sander Grefte
- Human and Animal Physiology, Wageningen University, Wageningen, the Netherlands
| | - Guido Jej Hooiveld
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands; Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
2
|
Karanfil AS, Louis F, Sowa Y, Matsusaki M. Polyelectrolyte nanofilms on cell surface can induce brown adipogenic differentiation of DFATs. Biochem Biophys Res Commun 2024; 733:150432. [PMID: 39043001 DOI: 10.1016/j.bbrc.2024.150432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 07/19/2024] [Indexed: 07/25/2024]
Abstract
Obesity and its related health issues significantly burden public health systems. Brown adipose tissue holds promise for addressing metabolic disorders and balancing the body's energy, making it a key research focus. Stimulating brown adipogenesis from stem cells could advance regenerative medicine and healthcare. In our previous research, we discovered that poly-l-lysine (PLL) significantly stimulates brown adipogenesis in three-dimensional differentiation of dedifferentiated fat cells (DFATs) within fibrin gels. In this study, we evaluated polyelectrolyte (PE) nanofilms made of PLL and dextran sulfate, applied directly to DFAT surfaces to improve brown adipogenic differentiation through an innovative approach. This approach involved coating the DFAT surfaces with PE nanofilms, forming a multilayer structure that not only provided a supportive matrix but also facilitated the adsorption of essential molecules like T3 and insulin for brown adipogenesis. DFATs coated with three PE layers and encapsulated in fibrin gel showed a significant increase in the adipogenic marker UCP1 gene expression and content. This PLL-based PE nanofilm coating on DFAT surfaces can be a novel and crucial technology for promoting brown adipogenesis in regenerative medicine and healthcare.
Collapse
Affiliation(s)
- Aslı Sena Karanfil
- Department of Applied Chemistry, Graduate School of Osaka University, Japan
| | - Fiona Louis
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Osaka University, Japan
| | - Yoshihiro Sowa
- Department of Plastic Surgery, Jichi Medical University, Shimotsuke, Tochigi Japan; Department of Plastic and Reconstructive Surgery, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Japan; Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Osaka University, Japan; Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Osaka University, Japan.
| |
Collapse
|
3
|
Reilly S, Aksu A, Zerbel Z, Dhillon P, Kelemen Y, Gbayisomore O, Chen S, Ahmadian M. Fatty acids promote uncoupled respiration via the ATP/ADP carrier in white adipocytes. RESEARCH SQUARE 2024:rs.3.rs-5094089. [PMID: 39399690 PMCID: PMC11469371 DOI: 10.21203/rs.3.rs-5094089/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Adipocytes store energy as triglycerides, while mobilizing energy when needed via lipolysis. Triglyceride lipolysis releases fatty acids and glycerol into the circulation to fuel other tissues. However, a significant fraction of fatty acids released by lipolysis are retained within the white adipose tissue and handled by adipocytes. While some of these retained fatty acids are re-esterified in white adipocytes1-6, the a substantial amount undergo oxidative metabolism via a pathway regulated by the nongenomic effects of STAT37-10. Here we report that fatty acids promote uncoupled oxidative metabolism in white adipocytes via the ATP/ADP carrier, contributing to thermogenesis and cold tolerance in obese thermoneutral-adapted mice, independent of brown adipose tissue and muscle activity. Our results suggest that uncoupled respiration in white adipocytes significantly contributes to whole-body energy expenditure and could be a promising target for obesity treatment.
Collapse
Affiliation(s)
| | - Ayla Aksu
- Weill Medical College of Cornell University
| | | | | | | | | | | | | |
Collapse
|
4
|
Reyad-Ul-Ferdous M, Gul I, Raheem MA, Pandey V, Qin P. Mitochondrial UCP1: Potential thermogenic mechanistic switch for the treatment of obesity and neurodegenerative diseases using natural and epigenetic drug candidates. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155672. [PMID: 38810549 DOI: 10.1016/j.phymed.2024.155672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/28/2024] [Accepted: 04/21/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND Brown fat is known to provide non-shivering thermogenesis through mitochondrial uncoupling mediated by uncoupling protein 1 (UCP1). Non-shivering is not dependent on UCP2, UCP4, and BMCP1/UCP5 genes, which are distinct from UCP1 in a way that they are not constitutive uncouplers. Although they are susceptible to free fatty acid and free radical activation, their functioning has a significant impact on the performance of neurons. METHODOLOGY Using subject-specific keywords (Adipose tissue; Adipocytes; Mitochondria; Obesity; Thermogenesis; UCP's in Neurodegeneration; Alzheimer's disease; Parkinson's disease), research articles and reviews were retrieved from Web of Science, ScienceDirect, Google Scholar, and PubMed. This article includespublications published between 2018 and 2023. The drugs that upregulate UCP1 are included in the study while the drugs that do not impact UCP1 are were not included. RESULTS Neuronal UCPs have a direct impact on synaptic plasticity, neurodegenerative processes, and neurotransmission, by modulating calcium flux, mitochondrial biogenesis, local temperature, and free radical generation. Numerous significant advances in the study of neuronal UCPs and neuroprotection are still to be made. Identification of the tissue-dependent effects of UCPs is essential first. Pharmacologically targeting neuronal UCPs is a key strategy for preventing both neurodegenerative diseases and physiological aging. Given that UCP2 has activities that are tissue-specific, it will be essential to develop treatments without harmful side effects. The triggering of UCPs by CoQ, an essential cofactor, produces nigral mitochondrial uncoupling, reduces MPTP-induced toxicity, and may even decrease the course of Parkinson's disease, according to early indications. CONCLUSION Herein, we explore the potential of UCP1 as a therapeutic target for treating obesity, neurodegenerative diseases as well as a potential activator of both synthetic and natural drugs. A deeper knowledge of synaptic signaling and neurodegeneration may pave the way to new discoveries regarding the functioning and controlling of these genes.
Collapse
Affiliation(s)
- Md Reyad-Ul-Ferdous
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China.
| | - Ijaz Gul
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Muhammad Akmal Raheem
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Vijay Pandey
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Peiwu Qin
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| |
Collapse
|
5
|
Hoogstraten CA, Schirris TJJ, Russel FGM. Unlocking mitochondrial drug targets: The importance of mitochondrial transport proteins. Acta Physiol (Oxf) 2024; 240:e14150. [PMID: 38666512 DOI: 10.1111/apha.14150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 03/02/2024] [Accepted: 04/12/2024] [Indexed: 05/12/2024]
Abstract
A disturbed mitochondrial function contributes to the pathology of many common diseases. These organelles are therefore important therapeutic targets. On the contrary, many adverse effects of drugs can be explained by a mitochondrial off-target effect, in particular, due to an interaction with carrier proteins in the inner membrane. Yet this class of transport proteins remains underappreciated and understudied. The aim of this review is to provide a deeper understanding of the role of mitochondrial carriers in health and disease and their significance as drug targets. We present literature-based evidence that mitochondrial carrier proteins are associated with prevalent diseases and emphasize their potential as drug (off-)target sites by summarizing known mitochondrial drug-transporter interactions. Studying these carriers will enhance our knowledge of mitochondrial drug on- and off-targets and provide opportunities to further improve the efficacy and safety of drugs.
Collapse
Affiliation(s)
- Charlotte A Hoogstraten
- Department of Pharmacy, Division of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, the Netherlands
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Tom J J Schirris
- Department of Pharmacy, Division of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, the Netherlands
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Frans G M Russel
- Department of Pharmacy, Division of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, the Netherlands
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
6
|
Jacobsen L, Lydersen L, Khandelia H. ATP-Bound State of the Uncoupling Protein 1 (UCP1) from Molecular Simulations. J Phys Chem B 2023; 127:9685-9696. [PMID: 37921649 DOI: 10.1021/acs.jpcb.3c03473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
The uncoupling protein 1 (UCP1) dissipates the transmembrane (TM) proton gradient in the inner mitochondrial membrane (IMM) by leaking protons across the membrane and producing heat in the process. Such a nonshivering production of heat in the brown adipose tissue can combat obesity-related diseases. UCP1-associated proton leak is activated by free fatty acids and inhibited by purine nucleotides. The mechanism of proton leak and the binding sites of the activators (fatty acids) remain unknown, while the binding site of the inhibitors (nucleotides) was described recently. Using molecular dynamics simulations, we generated a conformational ensemble of UCP1. Using metadynamics-based free energy calculations, we obtained the most likely ATP-bound conformation of UCP1. Our conformational ensemble provides a molecular basis for a breadth of prior biochemical data available for UCP1. Based on the simulations, we make the following testable predictions about the mechanisms of activation of proton leak and proton leak inhibition by ATP: (1) R277 plays the dual role of stabilizing ATP at the binding site for inhibition and acting as a proton surrogate for D28 in the absence of a proton during proton transport, (2) the binding of ATP to UCP1 is mediated by residues R84, R92, R183, and S88, (3) R92 shuttles ATP from the E191-R92 gate in the intermembrane space to the nucleotide binding site and serves to increase ATP affinity, (4) ATP can inhibit proton leak by controlling the ionization states of matrix facing lysine residues such as K269 and K56, and (5) fatty acids can bind to UCP1 from the IMM either via the cavity between TM1 and TM2 or between TM5 and TM6. Our simulations set the platform for future investigations into the proton transport and inhibition mechanisms of UCP1.
Collapse
Affiliation(s)
- Luise Jacobsen
- PhyLife: Physical Life Science, Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Laura Lydersen
- PhyLife: Physical Life Science, Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Himanshu Khandelia
- PhyLife: Physical Life Science, Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| |
Collapse
|
7
|
Cero C, Shu W, Reese AL, Douglas D, Maddox M, Singh AP, Ali SL, Zhu AR, Katz JM, Pierce AE, Long KT, Nilubol N, Cypess RH, Jacobs JL, Tian F, Cypess AM. Standardized In Vitro Models of Human Adipose Tissue Reveal Metabolic Flexibility in Brown Adipocyte Thermogenesis. Endocrinology 2023; 164:bqad161. [PMID: 37944134 PMCID: PMC11032247 DOI: 10.1210/endocr/bqad161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 10/10/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
Functional human brown and white adipose tissue (BAT and WAT) are vital for thermoregulation and nutritional homeostasis, while obesity and other stressors lead, respectively, to cold intolerance and metabolic disease. Understanding BAT and WAT physiology and dysfunction necessitates clinical trials complemented by mechanistic experiments at the cellular level. These require standardized in vitro models, currently lacking, that establish references for gene expression and function. We generated and characterized a pair of immortalized, clonal human brown (hBA) and white (hWA) preadipocytes derived from the perirenal and subcutaneous depots, respectively, of a 40-year-old male individual. Cells were immortalized with hTERT and confirmed to be of a mesenchymal, nonhematopoietic lineage based on fluorescence-activated cell sorting and DNA barcoding. Functional assessments showed that the hWA and hBA phenocopied primary adipocytes in terms of adrenergic signaling, lipolysis, and thermogenesis. Compared to hWA, hBA were metabolically distinct, with higher rates of glucose uptake and lactate metabolism, and greater basal, maximal, and nonmitochondrial respiration, providing a mechanistic explanation for the association between obesity and BAT dysfunction. The hBA also responded to the stress of maximal respiration by using both endogenous and exogenous fatty acids. In contrast to certain mouse models, hBA adrenergic thermogenesis was mediated by several mechanisms, not principally via uncoupling protein 1 (UCP1). Transcriptomics via RNA-seq were consistent with the functional studies and established a molecular signature for each cell type before and after differentiation. These standardized cells are anticipated to become a common resource for future physiological, pharmacological, and genetic studies of human adipocytes.
Collapse
Affiliation(s)
- Cheryl Cero
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Weiguo Shu
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Amy L Reese
- American Type Culture Collection, Sequencing and Bioinformatics Center, 10801 University Blvd, Manassas, VA 20110, USA
| | - Diana Douglas
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Michael Maddox
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
- Current Affiliation: Vita Therapeutics, 801 W. Baltimore Street, Suite 301, Baltimore, MD 21201, USA
| | - Ajeet P Singh
- American Type Culture Collection, Sequencing and Bioinformatics Center, 10801 University Blvd, Manassas, VA 20110, USA
| | - Sahara L Ali
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alexander R Zhu
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jacqueline M Katz
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anne E Pierce
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kelly T Long
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Naris Nilubol
- Surgical Oncology Program, Center for Cancer Research, NCI, NIH, 10 Center Drive, Room 4-5952, Bethesda, MD 20892, USA
| | - Raymond H Cypess
- American Type Culture Collection, 10801 University Blvd, Manassas, VA 20110, USA
| | - Jonathan L Jacobs
- American Type Culture Collection, Sequencing and Bioinformatics Center, 10801 University Blvd, Manassas, VA 20110, USA
| | - Fang Tian
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
8
|
Abstract
Recent advances in pharmacotherapies that promote appetite suppression have shown remarkable weight loss. Therapies targeting energy expenditure lag behind, and as such none have yet been identified to be safe and efficacious for sustaining negative energy balance toward weight loss. Multiple energy dissipating pathways have been identified in adipose tissue and muscle. The molecular effectors of some of these pathways have been identified, but much is still left to be learned about their regulation. Understanding the molecular underpinnings of metabolic inefficiency in adipose tissue and muscle is required if these pathways are to be therapeutically targeted in the context of obesity and obesity-accelerated diseases.
Collapse
Affiliation(s)
- Lawrence Kazak
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| |
Collapse
|
9
|
Jagtap U, Paul A. UCP1 activation: Hottest target in the thermogenesis pathway to treat obesity using molecules of synthetic and natural origin. Drug Discov Today 2023; 28:103717. [PMID: 37467882 DOI: 10.1016/j.drudis.2023.103717] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/03/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023]
Abstract
Uncoupling protein 1 (UCP1) has been discovered as a possible target for obesity treatment because of its widespread distribution in the inner mitochondrial membrane of brown adipose tissue (BAT) and high energy expenditure capabilities to burn calories as heat. UCP1 is dormant and does not produce heat without activation as it is inhibited by purine nucleotides. However, activation of UCP1 via either direct interaction with the UCP1 protein, an increase in the expression of UCP1 genes or the physiological production of fatty acids can lead to a rise in the thermogenesis phenomenon. Hence, activation of UCP1 through small molecules of synthetic and natural origin can be considered as a promising strategy to mitigate obesity.
Collapse
Affiliation(s)
- Utkarsh Jagtap
- Laboratory of Natural Product Chemistry, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Atish Paul
- Laboratory of Natural Product Chemistry, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India.
| |
Collapse
|
10
|
Shu H, Zhang J, Cheng D, Zhao X, Ma Y, Zhang C, Zhang Y, Jia Z, Liu Z. The Role of Proton-Coupled Amino Acid Transporter 2 (SLC36A2) in Cold-Induced Thermogenesis of Mice. Nutrients 2023; 15:3552. [PMID: 37630739 PMCID: PMC10458080 DOI: 10.3390/nu15163552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Brown adipocytes mainly utilize glucose and fatty acids to produce energy, which play key roles in thermogenesis. Furthermore, brown adipocytes also utilize other substrates, such as amino acids, for energy expenditure in various conditions. Here, we report the new physiological roles of proton-coupled amino acid transporters, SLC36A2 and SLC36A3, on global energy metabolism. The relative mRNA expression levels of both Slc36a2 and Slc36a3 were all highest in brown adipose tissue. We then generated global Slc36a2 and Slc36a3 knockout mice to investigate their functions in metabolism. Neither loss of Slc36a2 nor Slc36a3 affected the body weight and body composition of the mice. Slc36a2 knockout mice exhibited increased oxygen consumption during the daytime. After cold treatment, inhibition of Slc36a2 significantly decreased the mass of brown adipose tissue compared to wildtype mice, while it lowered the expression level of Cpt1a. Moreover, the serum lipid levels and liver mass were also decreased in Slc36a2 knockout mice after cold treatment. On the contrary, Slc36a3 knockout impaired glucose tolerance and up-regulated serum LDL-cholesterol concentration. Thus, SLC36A2 and SLC36A3 play central and different roles in the energy metabolism of the mice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhihao Jia
- Cambridge-Suda Genomic Resource Center, Suzhou Medical College, Soochow University, Suzhou 215123, China; (H.S.); (J.Z.); (Y.M.); (Y.Z.)
| | - Zhiwei Liu
- Cambridge-Suda Genomic Resource Center, Suzhou Medical College, Soochow University, Suzhou 215123, China; (H.S.); (J.Z.); (Y.M.); (Y.Z.)
| |
Collapse
|
11
|
Karanfil AS, Louis F, Matsusaki M. Biofabrication of vascularized adipose tissues and their biomedical applications. MATERIALS HORIZONS 2023; 10:1539-1558. [PMID: 36789675 DOI: 10.1039/d2mh01391f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Recent advances in adipose tissue engineering and cell biology have led to the development of innovative therapeutic strategies in regenerative medicine for adipose tissue reconstruction. To date, the many in vitro and in vivo models developed for vascularized adipose tissue engineering cover a wide range of research areas, including studies with cells of various origins and types, polymeric scaffolds of natural and synthetic derivation, models presented using decellularized tissues, and scaffold-free approaches. In this review, studies on adipose tissue types with different functions, characteristics and body locations have been summarized with 3D in vitro fabrication approaches. The reason for the particular focus on vascularized adipose tissue models is that current liposuction and fat transplantation methods are unsuitable for adipose tissue reconstruction as the lack of blood vessels results in inadequate nutrient and oxygen delivery, leading to necrosis in situ. In the first part of this paper, current studies and applications of white and brown adipose tissues are presented according to the polymeric materials used, focusing on the studies which could show vasculature in vitro and after in vivo implantation, and then the research on adipose tissue fabrication and applications are explained.
Collapse
Affiliation(s)
- Aslı Sena Karanfil
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan.
| | - Fiona Louis
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan.
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
| |
Collapse
|
12
|
Oflaz FE, Koshenov Z, Hirtl M, Bachkoenig OA, Graier WF, Gottschalk B. Synergy of Uncoupling Proteins (1 and 2) with Mitochondrial Ca2+ Uptake Machinery Potentiate Mitochondrial Uncoupling. Cell Calcium 2023; 112:102736. [PMID: 37031662 DOI: 10.1016/j.ceca.2023.102736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/30/2023] [Accepted: 04/04/2023] [Indexed: 04/07/2023]
Abstract
Mitochondrial uncoupling proteins UCP1 and UCP2 have a structural homology of app. 60%. They execute their mitochondria uncoupling function through different molecular mechanisms. Non-shivering thermogenesis by UCP1 is mediated through a transmembrane dissipation of the proton motive force to create heat during sympathetic stimulation. UCP2, on the other hand, modulates through the interaction with methylated MICU1 the permeability of the cristae junction, which acts as an isolator for the cristae-located mitochondrial membrane potential. In this mini-review, we discuss and compare the recently described molecular mechanism of UCP1 in brown adipose tissue and UCP2 in aged and cancer non-excitable cells that contribute to mitochondrial uncoupling, and the synergistic effects of both UCPs with the mitochondrial Ca2+ uptake machinery.
Collapse
Affiliation(s)
- Furkan E Oflaz
- Gottfried Schatz Research Center: Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, Graz, 8010 Austria
| | - Zhanat Koshenov
- Gottfried Schatz Research Center: Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, Graz, 8010 Austria
| | - Martin Hirtl
- Gottfried Schatz Research Center: Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, Graz, 8010 Austria
| | - Olaf A Bachkoenig
- Gottfried Schatz Research Center: Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, Graz, 8010 Austria
| | - Wolfgang F Graier
- Gottfried Schatz Research Center: Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, Graz, 8010 Austria; BioTechMed, Graz, Austria.
| | - Benjamin Gottschalk
- Gottfried Schatz Research Center: Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/4, Graz, 8010 Austria
| |
Collapse
|
13
|
Chang JS. Recent insights into the molecular mechanisms of simultaneous fatty acid oxidation and synthesis in brown adipocytes. Front Endocrinol (Lausanne) 2023; 14:1106544. [PMID: 36896177 PMCID: PMC9989468 DOI: 10.3389/fendo.2023.1106544] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/10/2023] [Indexed: 02/25/2023] Open
Abstract
Brown adipocytes is a specialized fat cell that dissipates nutrient-derived chemical energy in the form of heat, instead of ATP synthesis. This unique feature provides a marked capacity for brown adipocyte mitochondria to oxidize substrates independent of ADP availability. Upon cold exposure, brown adipocytes preferentially oxidize free fatty acids (FFA) liberated from triacylglycerol (TAG) in lipid droplets to support thermogenesis. In addition, brown adipocytes take up large amounts of circulating glucose, concurrently increasing glycolysis and de novo FA synthesis from glucose. Given that FA oxidation and glucose-derived FA synthesis are two antagonistic mitochondrial processes in the same cell, it has long been questioned how brown adipocytes run FA oxidation and FA synthesis simultaneously. In this review, I summarize mechanisms regulating mitochondrial substrate selection and describe recent findings of two distinct populations of brown adipocyte mitochondria with different substrate preferences. I further discuss how these mechanisms may permit a concurrent increase in glycolysis, FA synthesis, and FA oxidation in brown adipocytes.
Collapse
|
14
|
Varanita T, Angi B, Scattolini V, Szabo I. Kv1.3 K + Channel Physiology Assessed by Genetic and Pharmacological Modulation. Physiology (Bethesda) 2023; 38:0. [PMID: 35998249 DOI: 10.1152/physiol.00010.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Potassium channels are widespread over all kingdoms and play an important role in the maintenance of cellular ionic homeostasis. Kv1.3 is a voltage-gated potassium channel of the Shaker family with a wide tissue expression and a well-defined pharmacology. In recent decades, experiments mainly based on pharmacological modulation of Kv1.3 have highlighted its crucial contribution to different fundamental processes such as regulation of proliferation, apoptosis, and metabolism. These findings link channel function to various pathologies ranging from autoimmune diseases to obesity and cancer. In the present review, we briefly summarize studies employing Kv1.3 knockout animal models to confirm such roles and discuss the findings in comparison to the results obtained by pharmacological modulation of Kv1.3 in various pathophysiological settings. We also underline how these studies contributed to our understanding of channel function in vivo and propose possible future directions.
Collapse
Affiliation(s)
| | - Beatrice Angi
- Department of Biology, University of Padova, Padova, Italy
| | | | - Ildiko Szabo
- Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
15
|
Snoke DB, Mahler CA, Angelotti A, Cole RM, Sparagna GC, Baskin KK, Belury MA. Linoleic Acid-Enriched Diet Increases Mitochondrial Tetralinoleoyl Cardiolipin, OXPHOS Protein Levels, and Uncoupling in Interscapular Brown Adipose Tissue during Diet-Induced Weight Gain. BIOLOGY 2022; 12:9. [PMID: 36671702 PMCID: PMC9854875 DOI: 10.3390/biology12010009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/02/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
Cardiolipin (CL) is a phospholipid unique to the inner mitochondrial membrane that supports respiratory chain structure and function and is demonstrated to be influenced by types of dietary fats. However, the influence of dietary fat on CL species and how this best supports mitochondrial function in brown adipose tissue (BAT), which exhibits an alternative method of energy utilization through the uncoupling of the mitochondrial proton gradient to generate heat, is not well understood. Therefore, the aim of our study was to evaluate metabolic parameters, interscapular BAT CL quantity, species, and mitochondrial function in mice consuming isocaloric moderate-fat diets with either lard (LD; similar fatty acid profile to western dietary patterns) or safflower oil high in linoleic acid (SO), shown to be metabolically favorable in large clinical meta-analyses. Mice fed the SO diet exhibited decreased adiposity, improved insulin sensitivity, and enrichment of LA-containing CL species in BAT CL. Furthermore, mice fed the SO diet exhibit higher levels of OXPHOS complex proteins and increased oxygen consumption in BAT. Our findings demonstrate that dietary consumption of LA-rich oil improves metabolic parameters, increases LA-containing CL species, and improves BAT function when compared to the consumption of lard in mice during diet-induced weight gain.
Collapse
Affiliation(s)
- Deena B. Snoke
- Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
- Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, OH 43210, USA
| | - Connor A. Mahler
- Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, OH 43210, USA
- Indiana Biosciences Research Institute, Indianapolis, IN 46062, USA
| | - Austin Angelotti
- Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, OH 43210, USA
| | - Rachel M. Cole
- Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, OH 43210, USA
| | - Genevieve C. Sparagna
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kedryn K. Baskin
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Martha A. Belury
- Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
16
|
Xue K, Wu D, Wang Y, Zhao Y, Shen H, Yao J, Huang X, Li X, Zhou Z, Wang Z, Qiu Y. The mitochondrial calcium uniporter engages UCP1 to form a thermoporter that promotes thermogenesis. Cell Metab 2022; 34:1325-1341.e6. [PMID: 35977541 DOI: 10.1016/j.cmet.2022.07.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 03/03/2022] [Accepted: 07/25/2022] [Indexed: 01/08/2023]
Abstract
Uncoupling protein 1 (UCP1)-mediated adaptive thermogenesis protects mammals against hypothermia and metabolic dysregulation. Whether and how mitochondrial calcium regulates this process remains unclear. Here, we show that mitochondrial calcium uniporter (MCU) recruits UCP1 through essential MCU regulator (EMRE) to form an MCU-EMRE-UCP1 complex upon adrenergic stimulation. This complex formation increases mitochondrial calcium uptake to accelerate the tricarboxylic acid cycle and supply more protons that promote uncoupled respiration, functioning as a thermogenic uniporter. Mitochondrial calcium uptake 1 (MICU1) negatively regulates thermogenesis probably through inhibiting thermogenic uniporter formation. Accordingly, the deletion of Mcu or Emre in brown adipocytes markedly impairs thermogenesis and exacerbates obesity and metabolic dysfunction. Remarkably, the enhanced assembly of the thermogenic uniporter via Micu1 knockout or expressing linked EMRE-UCP1 results in opposite phenotypes. Thus, we have uncovered a "thermoporter" that provides a driving force for the UCP1 operation in thermogenesis, which could be leveraged to combat obesity and associated metabolic disorders.
Collapse
Affiliation(s)
- Kaili Xue
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Dongmei Wu
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Yushuang Wang
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Yiheng Zhao
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Hongyu Shen
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jingfei Yao
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Xun Huang
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Xinmeng Li
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Zhao Zhou
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Zihao Wang
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Yifu Qiu
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
17
|
Zhou D, Zhuan Q, Luo Y, Liu H, Meng L, Du X, Wu G, Hou Y, Li J, Fu X. Mito-Q promotes porcine oocytes maturation by maintaining mitochondrial thermogenesis via UCP2 downregulation. Theriogenology 2022; 187:205-214. [PMID: 35644089 DOI: 10.1016/j.theriogenology.2022.05.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/02/2022] [Accepted: 05/11/2022] [Indexed: 12/30/2022]
Abstract
Mitochondrial thermogenesis is an adaptive response of cells to their surrounding stress. Oxidative stress is one of the common stresses during in vitro maturation (IVM) of oocytes, which leads to mitochondrial dysfunction. This study aimed to probe the effects of the mitochondria-targeted antioxidant Mito-Q on oocyte development and unravel the role of Mito-Q in mitochondrial ATP production and thermogenesis regulation. Our results showed that Mito-Q had a positive effect on porcine oocytes maturation and subsequent embryo development. During oocytes IVM, Mito-Q could reduce ATP levels and ROS, increase lipid droplets accumulation, induce autophagy, and maintain mitochondrial temperature stability. Moreover, in metaphase II (MII) oocytes, Mito-Q would induce mitochondrial uncoupling manifested by decreased ATP, attenuated mitochondrial membrane potential (MMP), and increased mitochondrial thermogenesis. Notably, the expression of mitochondrial uncoupling protein (UCP2) was significantly reduced in oocytes treated with Mito-Q. Further study indicated that specific depletion of UCP2 in oocytes also resulted in increased thermogenesis, decreased ATP and declined MMP, suggesting that UCP2 downregulation may participate in Mito-Q-induced mitochondrial uncoupling. In summary, our data demonstrate that Mito-Q promotes oocyte maturation in vitro and maintains the stability of mitochondrial thermogenesis by inhibiting UCP2 expression.
Collapse
Affiliation(s)
- Dan Zhou
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science, Beijing, 100193, China
| | - Qingrui Zhuan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science, Beijing, 100193, China
| | - Yuwen Luo
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science, Beijing, 100193, China
| | - Hongyu Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science, Beijing, 100193, China
| | - Lin Meng
- State Key Laboratories of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Xingzhu Du
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science, Beijing, 100193, China
| | - Guoquan Wu
- Yunnan Provincial Engineering Laboratory of Animal Genetic Resource Conservation and Germplasm Enhancement, Yunnan Animal Science and Veterinary Institute, Kunming, Yunnan, 650224, China
| | - Yunpeng Hou
- State Key Laboratories of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Jun Li
- Department of Reproductive Medicine, Reproductive Medical Center, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050031, China.
| | - Xiangwei Fu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science, Beijing, 100193, China; State Key Laboratory of Sheep Genetic Improvement and Healthy Breeding, Xinjiang Academy of Agricultural and Reclamation Sciences, Shihezi, Xinjiang, 832000, China.
| |
Collapse
|
18
|
Horino M, Ikeda K, Yamada T. The Role of Thermogenic Fat Tissue in Energy Consumption. Curr Issues Mol Biol 2022; 44:3166-3179. [PMID: 35877443 PMCID: PMC9317885 DOI: 10.3390/cimb44070219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 12/19/2022] Open
Abstract
Mammalian adipose tissues are broadly divided into white adipose tissue (WAT) and thermogenic fat tissue (brown adipose tissue and beige adipose tissue). Uncoupling protein 1 (UCP1) is the central protein in thermogenesis, and cells that exhibit induced UCP1 expression and appear scattered throughout WAT are called beige adipocytes, and their induction in WAT is referred to as “beiging”. Beige adipocytes can differentiate from preadipocytes or convert from mature adipocytes. UCP1 was thought to contribute to non-shivering thermogenesis; however, recent studies demonstrated the presence of UCP1-independent thermogenic mechanisms. There is evidence that thermogenic fat tissue contributes to systemic energy expenditure even in human beings. This review discusses the roles that thermogenic fat tissue plays in energy consumption and offers insight into the possibility and challenges associated with its application in the treatment of obesity and type 2 diabetes.
Collapse
|
19
|
Bertholet AM, Natale AM, Bisignano P, Suzuki J, Fedorenko A, Hamilton J, Brustovetsky T, Kazak L, Garrity R, Chouchani ET, Brustovetsky N, Grabe M, Kirichok Y. Mitochondrial uncouplers induce proton leak by activating AAC and UCP1. Nature 2022; 606:180-187. [PMID: 35614225 PMCID: PMC9646675 DOI: 10.1038/s41586-022-04747-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/08/2022] [Indexed: 11/08/2022]
Abstract
Mitochondria generate heat due to H+ leak (IH) across their inner membrane1. IH results from the action of long-chain fatty acids on uncoupling protein 1 (UCP1) in brown fat2-6 and ADP/ATP carrier (AAC) in other tissues1,7-9, but the underlying mechanism is poorly understood. As evidence of pharmacological activators of IH through UCP1 and AAC is lacking, IH is induced by protonophores such as 2,4-dinitrophenol (DNP) and cyanide-4-(trifluoromethoxy) phenylhydrazone (FCCP)10,11. Although protonophores show potential in combating obesity, diabetes and fatty liver in animal models12-14, their clinical potential for treating human disease is limited due to indiscriminately increasing H+ conductance across all biological membranes10,11 and adverse side effects15. Here we report the direct measurement of IH induced by DNP, FCCP and other common protonophores and find that it is dependent on AAC and UCP1. Using molecular structures of AAC, we perform a computational analysis to determine the binding sites for protonophores and long-chain fatty acids, and find that they overlap with the putative ADP/ATP-binding site. We also develop a mathematical model that proposes a mechanism of uncoupler-dependent IH through AAC. Thus, common protonophoric uncouplers are synthetic activators of IH through AAC and UCP1, paving the way for the development of new and more specific activators of these two central mediators of mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Ambre M Bertholet
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Andrew M Natale
- Cardiovascular Research Institute, Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Paola Bisignano
- Cardiovascular Research Institute, Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Junji Suzuki
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Andriy Fedorenko
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - James Hamilton
- Department of Pharmacology and Toxicology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Tatiana Brustovetsky
- Department of Pharmacology and Toxicology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Lawrence Kazak
- Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Ryan Garrity
- Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Edward T Chouchani
- Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Nickolay Brustovetsky
- Department of Pharmacology and Toxicology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Michael Grabe
- Cardiovascular Research Institute, Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA.
| | - Yuriy Kirichok
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
20
|
Lytic cocktail: An effective method to alleviate severe burn induced hyper-metabolism through regulating white adipose tissue browning. Heliyon 2022; 8:e09128. [PMID: 35846468 PMCID: PMC9280373 DOI: 10.1016/j.heliyon.2022.e09128] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/28/2021] [Accepted: 03/14/2022] [Indexed: 11/10/2022] Open
Abstract
Background Browning of white adipose tissue is associated with elevated resting metabolic rates and is considered to be one of the indispensable causes of hypermetabolism in burn patients. Hypermetabolism means increased resting energy expenditure, raised body temperature and acute-phase proteins. Persistently elevated levels of circulating stress hormones have been reported to induce browning of subcutaneous white adipose tissue. The lytic cocktail is a combination of medicines pethidine, chlorpromazine, and promethazine that has been used clinically in sedation for the management of patients. As reported this sedative treatment can reduce the expression of catecholamines in major burn rats. Thus, in this paper we focused on the effects of lytic cocktail in the regulation of white adipose tissue browning and hypermetabolism and we further investigated the underlying mechanism. Methods A 30% total body surface area (TBSA) Ⅲ degree scald rat model was used for this study. The rats were randomly divided into a sham scald group, a scalding with immediate resuscitation group, and a group of scalding with immediate resuscitation and lytic cocktail treatment. The levels of norepinephrine and epinephrine in plasma were dynamically detected. Changes of the rat body weight and food intake were recorded and compared as indexes of metabolism responses after post-scalding. For the study of white adipose tissue browning, inguinal adipose tissue was used. Metabolic changes, while indicatives of white fat browning were measured by PET/CT. The expression of white adipose browning related proteins and the changes of mitochondria number were used to assess browning of inguinal adipose. Results The level of plasma catecholamines norepinephrine and epinephrine in the lytic cocktail-treated group was significantly lower than the other two groups. Morphology and PET/CT showed that the inguinal white adipose browning was inhibited in the lytic cocktail treated group, whereas scalding with immediate resuscitation group showed browning of white adipose. The number of mitochondria, the expressions of white adipose browning related proteins in the lytic cocktail group were also significantly lower than that of the group of scalding with immediate resuscitation. Conclusion By reducing expression of heat-related proteins, the application of lytic cocktail medicines inhibits the white adipose tissue browning, which suppresses hypermetabolism in scalded rats. The mechanism might be related to decreased expression levels of stress hormones induced by lytic cocktail. This research suggests that lytic cocktails may be an effective treatment for hypermetabolism after severe burn injury.
Collapse
|
21
|
Abstract
Mitochondria of all tissues convert various metabolic substrates into two forms of energy: ATP and heat. Historically, the primary focus of research in mitochondrial bioenergetics was on the mechanisms of ATP production, while mitochondrial thermogenesis received significantly less attention. Nevertheless, mitochondrial heat production is crucial for the maintenance of body temperature, regulation of the pace of metabolism, and prevention of oxidative damage to mitochondria and the cell. In addition, mitochondrial thermogenesis has gained significance as a pharmacological target for treating metabolic disorders. Mitochondria produce heat as the result of H+ leak across their inner membrane. This review provides a critical assessment of the current field of mitochondrial H+ leak and thermogenesis, with a focus on the molecular mechanisms involved in the function and regulation of uncoupling protein 1 and the ADP/ATP carrier, the two proteins that mediate mitochondrial H+ leak.
Collapse
Affiliation(s)
- Ambre M. Bertholet
- Department of Physiology, University of California San Francisco, 600 16 Street, San Francisco, CA 94158, USA,Department of Physiology, David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, Los Angeles, CA 90095, USA,Corresponding authors: ,
| | - Yuriy Kirichok
- Department of Physiology, University of California, San Francisco, California, USA;
| |
Collapse
|
22
|
Sakers A, De Siqueira MK, Seale P, Villanueva CJ. Adipose-tissue plasticity in health and disease. Cell 2022; 185:419-446. [PMID: 35120662 PMCID: PMC11152570 DOI: 10.1016/j.cell.2021.12.016] [Citation(s) in RCA: 363] [Impact Index Per Article: 121.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/08/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022]
Abstract
Adipose tissue, colloquially known as "fat," is an extraordinarily flexible and heterogeneous organ. While historically viewed as a passive site for energy storage, we now appreciate that adipose tissue regulates many aspects of whole-body physiology, including food intake, maintenance of energy levels, insulin sensitivity, body temperature, and immune responses. A crucial property of adipose tissue is its high degree of plasticity. Physiologic stimuli induce dramatic alterations in adipose-tissue metabolism, structure, and phenotype to meet the needs of the organism. Limitations to this plasticity cause diminished or aberrant responses to physiologic cues and drive the progression of cardiometabolic disease along with other pathological consequences of obesity.
Collapse
Affiliation(s)
- Alexander Sakers
- Institute for Diabetes, Obesity & Metabolism, Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Mirian Krystel De Siqueira
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, Los Angeles, CA 90095-7070 USA; Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095-7070 USA
| | - Patrick Seale
- Institute for Diabetes, Obesity & Metabolism, Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104 USA.
| | - Claudio J Villanueva
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, Los Angeles, CA 90095-7070 USA; Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095-7070 USA.
| |
Collapse
|
23
|
Tang R, Sun W, Zhang JC, Mao L, Quanquin N, Wu D, Sun Y. Expression of Human Uncoupling Protein-1 in Escherichia coli Decreases its Survival Under Extremely Acidic Conditions. Curr Microbiol 2022; 79:77. [DOI: 10.1007/s00284-022-02762-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 01/08/2022] [Indexed: 11/03/2022]
|
24
|
Uncoupling Proteins and Regulated Proton Leak in Mitochondria. Int J Mol Sci 2022; 23:ijms23031528. [PMID: 35163451 PMCID: PMC8835771 DOI: 10.3390/ijms23031528] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/26/2022] [Indexed: 12/17/2022] Open
Abstract
Higher concentration of protons in the mitochondrial intermembrane space compared to the matrix results in an electrochemical potential causing the back flux of protons to the matrix. This proton transport can take place through ATP synthase complex (leading to formation of ATP) or can occur via proton transporters of the mitochondrial carrier superfamily and/or membrane lipids. Some mitochondrial proton transporters, such as uncoupling proteins (UCPs), transport protons as their general regulating function; while others are symporters or antiporters, which use the proton gradient as a driving force to co-transport other substrates across the mitochondrial inner membrane (such as phosphate carrier, a symporter; or aspartate/glutamate transporter, an antiporter). Passage (or leakage) of protons across the inner membrane to matrix from any route other than ATP synthase negatively impacts ATP synthesis. The focus of this review is on regulated proton transport by UCPs. Recent findings on the structure and function of UCPs, and the related research methodologies, are also critically reviewed. Due to structural similarity of members of the mitochondrial carrier superfamily, several of the known structural features are potentially expandable to all members. Overall, this report provides a brief, yet comprehensive, overview of the current knowledge in the field.
Collapse
|
25
|
Liu X, Gao YP, Shen ZX, Qu YY, Liu WW, Yao D, Xing B, Xu ZH, Li X, Zhao QC. Study on the experimental verification and regulatory mechanism of Rougui-Ganjiang herb-pair for the actions of thermogenesis in brown adipose tissue based on network pharmacology. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:114378. [PMID: 34192599 DOI: 10.1016/j.jep.2021.114378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 06/25/2021] [Accepted: 06/25/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cinnamomum cassia Presl (Rougui) has character of xin、gan、wen, belongs to Jing of heart、lung、bladder, and has the effect of dispersing cold and relieving pain. It is widely used to resolve the exterior and dissipate cold in Treatise on Febrile Diseases (Shang Han Lun), such as Chaihu Guizhi Ganjiang Tang and Guizhi Renshen Tang. Both these two prescriptions contain Cinnamomum cassia Presl and Zingiber officinale Rosc (Ganjiang). Rougui-Ganjiang herb-pair (RGHP) can warm viscera and remove cold, which is widely used in Shang Han Lun. And in modern times, recent studies have showed that cinnamon and ginger also have the effect of thermogenesis and regulating the body temperature, respectively. AIM OF THE STUDY To maintain the body thermal homeostasis and prevent cold invasion of main organs, in this study, we assessed the underlying physiological changes induced by RGHP in mice exposed to -20 °C and explored the mechanisms for the thermogenic actions of RGHP in brown adipose tissue (BAT) by network pharmacology and molecular docking. MATERIALS AND METHODS Male Kunming (KM) mice were fed normal diet with orally administration of distilled water or ethanol RGHP extract (three doses: 375,750 and 1500 mg/kg) for 21 days, once per day and then exposed to -20 °C for 2 h. The core temperature, activity ability and the degree of frostbite in mice, morphological and ATP content of adipocytes were measured. In addition, the network pharmacology was employed to predict the targets of RGHP' s thermogenesis effect on BAT. Pathway analysis and biological process with key genes was carried out through KEGG and GO analysis, respectively. Furthermore, the core ingredients and targets obtained by network pharmacology were verified by molecular docking and Western blot assays. RESULTS RGHP can significantly increase the core body temperature, reduce the degree of frostbite and enhance the activity ability of mice after cold exposure. Meanwhile, it can also improve the lipid morphology and decrease ATP production in BAT. A network pharmacology-based analysis identified 246 ingredients from RGHP (two herbs), which related to 222 target genes. There were 8 common genes between 222 compounds target genes and 62 thermogenesis associated target genes, which linked to 49 potential compounds. There are 24 ingredients which degree are greater than the average. Among them, we found that oleic acid, EIC, 6-gingerol, eugenol, isohomogenol and sitogluside could be detected in mice plasma. The cAMP-PPAR signaling pathway was enriched for thermogenesis after KEGG analysis with 8 genes. Molecular docking analysis and Western blot assay further confirmed that oleic acid, 6-gingerol, eugenol and isohomogenol were potential active ingredients for RGHP's heat production effect. And UCP1, PGC-1α, PPARα and PPARγ are key thermogenesis proteins. CONCLUSIONS RGHP treatment can significantly maintain the rectal temperature of mice by enhancing the BAT heat production. RGHP exhibited the heat production effect, which might be mainly attributed to increasing thermogenesis through the cAMP-PPAR signaling pathway in cold exposure mice. Oleic acid, 6-gingerol, eugenol and isohomogenol might be considered the potential therapeutic ingredients which affect the key targets of thermogenesis effect.
Collapse
Affiliation(s)
- Xin Liu
- School of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, 110016, China; Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, China
| | - Ya-Ping Gao
- School of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Ze-Xu Shen
- School of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Ying-Ying Qu
- School of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Wen-Wu Liu
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Dong Yao
- College of Traditional Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, China
| | - Bo Xing
- School of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Zi-Hua Xu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, China
| | - Xiang Li
- School of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, 110016, China; Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, China.
| | - Qing-Chun Zhao
- School of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, 110016, China; Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, China.
| |
Collapse
|
26
|
Kharechkina ES, Nikiforova AB, Belosludtsev KN, Rokitskaya TI, Antonenko YN, Kruglov AG. Pioglitazone Is a Mild Carrier-Dependent Uncoupler of Oxidative Phosphorylation and a Modulator of Mitochondrial Permeability Transition. Pharmaceuticals (Basel) 2021; 14:ph14101045. [PMID: 34681269 PMCID: PMC8537895 DOI: 10.3390/ph14101045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 12/17/2022] Open
Abstract
Pioglitazone (PIO) is an insulin-sensitizing antidiabetic drug, which normalizes glucose and lipid metabolism but may provoke heart and liver failure and chronic kidney diseases. Both therapeutic and adverse effects of PIO can be accomplished through mitochondrial targets. Here, we explored the capability of PIO to modulate the mitochondrial membrane potential (ΔΨm) and the permeability transition pore (mPTP) opening in different models in vitro. ΔΨm was measured using tetraphenylphosphonium and the fluorescent dye rhodamine 123. The coupling of oxidative phosphorylation was estimated polarographically. The transport of ions and solutes across membranes was registered by potentiometric and spectral techniques. We found that PIO decreased ΔΨm in isolated mitochondria and intact thymocytes and the efficiency of ADP phosphorylation, particularly after the addition of Ca2+. The presence of the cytosolic fraction mitigated mitochondrial depolarization but made it sustained. Carboxyatractyloside diminished the PIO-dependent depolarization. PIO activated proton transport in deenergized mitochondria but not in artificial phospholipid vesicles. PIO had no effect on K+ and Ca2+ inward transport but drastically decreased the mitochondrial Ca2+-retention capacity and protective effects of adenine nucleotides against mPTP opening. Thus, PIO is a mild, partly ATP/ADP-translocase-dependent, uncoupler and a modulator of ATP production and mPTP sensitivity to Ca2+ and adenine nucleotides. These properties contribute to both therapeutic and adverse effects of PIO.
Collapse
Affiliation(s)
- Ekaterina S. Kharechkina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (E.S.K.); (A.B.N.); (K.N.B.)
| | - Anna B. Nikiforova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (E.S.K.); (A.B.N.); (K.N.B.)
| | - Konstantin N. Belosludtsev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (E.S.K.); (A.B.N.); (K.N.B.)
- Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia
| | - Tatyana I. Rokitskaya
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (T.I.R.); (Y.N.A.)
| | - Yuri N. Antonenko
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (T.I.R.); (Y.N.A.)
| | - Alexey G. Kruglov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (E.S.K.); (A.B.N.); (K.N.B.)
- Correspondence: ; Tel.: +7-4967-739107
| |
Collapse
|
27
|
Ardalan A, Sowlati-Hashjin S, Oduwoye H, Uwumarenogie SO, Karttunen M, Smith MD, Jelokhani-Niaraki M. Biphasic Proton Transport Mechanism for Uncoupling Proteins. J Phys Chem B 2021; 125:9130-9144. [PMID: 34365794 DOI: 10.1021/acs.jpcb.1c04766] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
It has been suggested that uncoupling proteins (UCPs) transport protons via interconversion between two conformational states: one in the "cytoplasmic state" and the other in the "matrix state". Matrix and cytoplasmic salt-bridge networks are key controllers of these states. This study proposes a mechanism for proton transport in tetrameric UCP2, with focus on the role of the matrix network. Eleven mutants were prepared to disrupt (K → Q or D → N mutations) or alter (K → D and D → K mutations) the salt-bridges in the matrix network. Proteins were recombinantly expressed in Escherichia coli membrane, reconstituted in model lipid membranes, and their structures and functions were analyzed by gel electrophoresis, circular dichroism spectroscopy, fluorescence assays, as well as molecular dynamics simulations. It is shown that the UCP2 matrix network contains five salt-bridges (rather than the previously reported three), and the matrix network can regulate the proton transport by holding the protein's transmembrane helices in close proximity, limiting the movement of the activator fatty acid(s). A biphasic two-state molecular model is proposed for proton transport in tetrameric (a dimer of stable dimers) UCP2, in which all the monomers are functional, and monomers in each dimer are in the same transport mode. Purine nucleotide (e.g., ATP) can occlude the internal pore of the monomeric units of UCP tetramers via interacting with positive residues at or in the proximity of the matrix network (K38, K141, K239, R88, R185, and R279) and prevent switching between cytoplasmic and matrix states, thus inhibiting the proton transport. This study provides new insights into the mechanism of proton transport and regulation in UCPs.
Collapse
Affiliation(s)
- Afshan Ardalan
- Department of Chemistry and Biochemistry, Wilfrid Laurier University, Waterloo, Ontario N2L 3C5, Canada
| | - Shahin Sowlati-Hashjin
- Department of Chemistry, The University of Western Ontario, London, Ontario N6A 3K7, Canada.,The Center for Advanced Materials and Biomaterials Research, The University of Western Ontario, London, Ontario N6K 3K7, Canada
| | - Habib Oduwoye
- Department of Chemistry and Biochemistry, Wilfrid Laurier University, Waterloo, Ontario N2L 3C5, Canada
| | - Stephanie O Uwumarenogie
- Department of Chemistry and Biochemistry, Wilfrid Laurier University, Waterloo, Ontario N2L 3C5, Canada
| | - Mikko Karttunen
- Department of Chemistry, The University of Western Ontario, London, Ontario N6A 3K7, Canada.,The Center for Advanced Materials and Biomaterials Research, The University of Western Ontario, London, Ontario N6K 3K7, Canada.,Department of Physics and Astronomy, The University of Western Ontario, London, Ontario N6A 3K7, Canada
| | - Matthew D Smith
- Department of Biology, Wilfrid Laurier University, Waterloo, Ontario N2L 3C5, Canada
| | - Masoud Jelokhani-Niaraki
- Department of Chemistry and Biochemistry, Wilfrid Laurier University, Waterloo, Ontario N2L 3C5, Canada
| |
Collapse
|
28
|
Lagarde D, Jeanson Y, Portais JC, Galinier A, Ader I, Casteilla L, Carrière A. Lactate Fluxes and Plasticity of Adipose Tissues: A Redox Perspective. Front Physiol 2021; 12:689747. [PMID: 34276410 PMCID: PMC8278056 DOI: 10.3389/fphys.2021.689747] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/09/2021] [Indexed: 12/18/2022] Open
Abstract
Lactate, a metabolite produced when the glycolytic flux exceeds mitochondrial oxidative capacities, is now viewed as a critical regulator of metabolism by acting as both a carbon and electron carrier and a signaling molecule between cells and tissues. In recent years, increasing evidence report its key role in white, beige, and brown adipose tissue biology, and highlights new mechanisms by which lactate participates in the maintenance of whole-body energy homeostasis. Lactate displays a wide range of biological effects in adipose cells not only through its binding to the membrane receptor but also through its transport and the subsequent effect on intracellular metabolism notably on redox balance. This study explores how lactate regulates adipocyte metabolism and plasticity by balancing intracellular redox state and by regulating specific signaling pathways. We also emphasized the contribution of adipose tissues to the regulation of systemic lactate metabolism, their roles in redox homeostasis, and related putative physiopathological repercussions associated with their decline in metabolic diseases and aging.
Collapse
Affiliation(s)
- Damien Lagarde
- Goodman Cancer Research Center, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada.,Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France
| | - Yannick Jeanson
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France
| | - Jean-Charles Portais
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France.,MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
| | - Anne Galinier
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France.,Institut Fédératif de Biologie, CHU Purpan, Toulouse, France
| | - Isabelle Ader
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France
| | - Louis Casteilla
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France
| | - Audrey Carrière
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France
| |
Collapse
|
29
|
Friedrich VK, Rubel MA, Schurr TG. Mitochondrial genetic variation in human bioenergetics, adaptation, and adult disease. Am J Hum Biol 2021; 34:e23629. [PMID: 34146380 DOI: 10.1002/ajhb.23629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES Mitochondria are critical for the survival of eukaryotic organisms due to their ability to produce cellular energy, which drives virtually all aspects of host biology. However, the effects of mitochondrial DNA (mtDNA) variation in relation to disease etiology and adaptation within contemporary global human populations remains incompletely understood. METHODS To develop a more holistic understanding of the role of mtDNA diversity in human adaptation, health, and disease, we investigated mitochondrial biology and bioenergetics. More specifically, we synthesized details from studies of mitochondrial function and variation in the context of haplogroup background, climatic adaptation, and oxidative disease. RESULTS The majority of studies show that mtDNA variation arose during modern human dispersal around the world. Some of these variants appear to have been positively selected for their adaptiveness in colder climates, with these sequence changes having implications for tissue-specific function and thermogenic capacity. In addition, many variants modulating energy production are also associated with damaging metabolic byproducts and mitochondrial dysfunction, which, in turn, are implicated in the onset and severity of several different adult mitochondrial diseases. Thus, mtDNA variation that governs bioenergetics, metabolism, and thermoregulation may potentially have adverse consequences for human health, depending on the genetic background and context in which it occurs. CONCLUSIONS Our review suggests that the mitochondrial research field would benefit from independently replicating mtDNA haplogroup-phenotype associations across global populations, incorporating potentially confounding environmental, demographic, and disease covariates into studies of mtDNA variation, and extending association-based studies to include analyses of complete mitogenomes and assays of mitochondrial function.
Collapse
Affiliation(s)
- Volney K Friedrich
- Department of Anthropology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Meagan A Rubel
- Department of Anthropology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Center for Translational Imaging and Precision Medicine, University of California - San Diego, La Jolla, California, USA
| | - Theodore G Schurr
- Department of Anthropology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
30
|
Camerino C. Oxytocin Involvement in Body Composition Unveils the True Identity of Oxytocin. Int J Mol Sci 2021; 22:ijms22126383. [PMID: 34203705 PMCID: PMC8232088 DOI: 10.3390/ijms22126383] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 01/11/2023] Open
Abstract
The origin of the Oxytocin/Vasopressin system dates back about 600 million years. Oxytocin (Oxt) together with Vasopressin (VP) regulate a diversity of physiological functions that are important for osmoregulation, reproduction, metabolism, and social behavior. Oxt/VP-like peptides have been identified in several invertebrate species and they are functionally related across the entire animal kingdom. Functional conservation enables future exploitation of invertebrate models to study Oxt’s functions not related to pregnancy and the basic mechanisms of central Oxt/VP signaling. Specifically, Oxt is well known for its effects on uteri contractility and milk ejection as well as on metabolism and energy homeostasis. Moreover, the striking evidence that Oxt is linked to energy regulation is that Oxt- and Oxytocin receptor (Oxtr)-deficient mice show late onset obesity. Interestingly Oxt−/− or Oxtr−/− mice develop weight gain without increasing food intake, suggesting that a lack of Oxt reduce metabolic rate. Oxt is expressed in a diversity of skeletal muscle phenotypes and regulates thermogenesis and bone mass. Oxt may increases skeletal muscle tonicity and/or increases body temperature. In this review, the author compared the three most recent theories on the effects of Oxt on body composition.
Collapse
Affiliation(s)
- Claudia Camerino
- Department of Biomedical Sciences and Human Oncology (Section of Pharmacology), School of Medicine, University of Bari Aldo Moro, P.za G. Cesare 11, 70100 Bari, Italy;
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy
| |
Collapse
|
31
|
Ramirez LA, Quezada J, Duarte L, Concha F, Escobillana L, Rincon-Cervera MA, Perez-Bravo F, Elorza AA, Bravo-Sagua R, Garcia-Diaz DF. The administration of an extract from Berberis microphylla stimulates energy expenditure, thermogenesis and mitochondrial dynamics in mice brown adipose tissue. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2021.100988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
32
|
Nicholls DG. Mitochondrial proton leaks and uncoupling proteins. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2021; 1862:148428. [PMID: 33798544 DOI: 10.1016/j.bbabio.2021.148428] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/15/2021] [Accepted: 03/25/2021] [Indexed: 01/02/2023]
Abstract
Non-shivering thermogenesis in brown adipose tissue is mediated by uncoupling protein 1 (UCP1), which provides a carefully regulated proton re-entry pathway across the mitochondrial inner membrane operating in parallel to the ATP synthase and allowing respiration, and hence thermogenesis, to be released from the constraints of respiratory control. In the 40 years since UCP1 was first described, an extensive, and frequently contradictory, literature has accumulated, focused on the acute physiological regulation of the protein by fatty acids, purine nucleotides and possible additional factors. The purpose of this review is to examine, in detail, the experimental evidence underlying these proposed mechanisms. Emphasis will be placed on the methodologies employed and their relation to the physiological constraints under which the protein functions in the intact cell. The nature of the endogenous, UCP1-independent, proton leak will also be discussed. Finally, the troubled history of the putative novel uncoupling proteins, UCP2 and UCP3, will be evaluated.
Collapse
|
33
|
Xiang-Li, Bo-Xing, Xin-Liu, Jiang XW, Lu HY, Xu ZH, Yue-Yang, Qiong-Wu, Dong-Yao, Zhang YS, Zhao QC. Network pharmacology-based research uncovers cold resistance and thermogenesis mechanism of Cinnamomum cassia. Fitoterapia 2021; 149:104824. [PMID: 33388379 DOI: 10.1016/j.fitote.2020.104824] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/25/2020] [Accepted: 12/26/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cinnamomum cassia (L.) J.Presl (Cinnamon) was known as a kind of hot herb, improved circulation and warmed the body. However, the active components and mechanisms of dispelling cold remain unknown. METHODS The effects of several Chinses herbs on thermogenesis were evaluated on body temperature and activation of brown adipose tissue. After confirming the effect, the components of cinnamon were identified using HPLC-Q-TOF/MS and screened with databases. The targets of components were obtained with TCMSP, SymMap, Swiss and STITCH databases. Thermogenesis genes were predicted with DisGeNET and GeneCards databases. The protein-protein interaction network was constructed with Cytoscape 3.7.1 software. GO enrichment analysis was accomplished with STRING databases. KEGG pathway analysis was established with Omicshare tools. The top 20 targets for four compounds were obtained according to the number of edges of PPI network. In addition, the network results were verified with experimental research for the effects of extracts and major compounds. RESULTS Cinnamon extract significantly upregulated the body temperature during cold exposure.121 components were identified in HPLC-Q-TOF/MS. Among them, 60 compounds were included in the databases. 116 targets were obtained for the compounds, and 41 genes were related to thermogenesis. The network results revealed that 27 active ingredients and 39 target genes. Through the KEGG analysis, the top 3 pathways were PPAR signaling pathway, AMPK signaling pathway, thermogenesis pathway. The thermogenic protein PPARγ, UCP1 and PGC1-α was included in the critical targets of four major compounds. The three major compounds increased the lipid consumption and activated the brown adipocyte. They also upregulated the expression of UCP1, PGC1-α and pHSL, especially 2-methoxycinnamaldehyde was confirmed the effect for the first time. Furthermore, cinnamaldehyde and cinnamon extract activated the expression of TRPA1 on DRG cells. CONCLUSION The mechanisms of cinnamon on cold resistance were investigated with network pharmacology and experiment validation. This work provided research direction to support the traditional applications of thermogenesis.
Collapse
Affiliation(s)
- Xiang-Li
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang 110016, China; Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, China
| | - Bo-Xing
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xin-Liu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiao-Wen Jiang
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hong-Yuan Lu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zi-Hua Xu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yue-Yang
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qiong-Wu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dong-Yao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, China
| | - Ying-Shi Zhang
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qing-Chun Zhao
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang 110016, China; Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, China.
| |
Collapse
|
34
|
Škulj S, Brkljača Z, Kreiter J, Pohl EE, Vazdar M. Molecular Dynamics Simulations of Mitochondrial Uncoupling Protein 2. Int J Mol Sci 2021; 22:ijms22031214. [PMID: 33530558 PMCID: PMC7866055 DOI: 10.3390/ijms22031214] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/19/2021] [Accepted: 01/22/2021] [Indexed: 11/16/2022] Open
Abstract
Molecular dynamics (MD) simulations of uncoupling proteins (UCP), a class of transmembrane proteins relevant for proton transport across inner mitochondrial membranes, represent a complicated task due to the lack of available structural data. In this work, we use a combination of homology modelling and subsequent microsecond molecular dynamics simulations of UCP2 in the DOPC phospholipid bilayer, starting from the structure of the mitochondrial ATP/ADP carrier (ANT) as a template. We show that this protocol leads to a structure that is impermeable to water, in contrast to MD simulations of UCP2 structures based on the experimental NMR structure. We also show that ATP binding in the UCP2 cavity is tight in the homology modelled structure of UCP2 in agreement with experimental observations. Finally, we corroborate our results with conductance measurements in model membranes, which further suggest that the UCP2 structure modeled from ANT protein possesses additional key functional elements, such as a fatty acid-binding site at the R60 region of the protein, directly related to the proton transport mechanism across inner mitochondrial membranes.
Collapse
Affiliation(s)
- Sanja Škulj
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička 54, 10000 Zagreb, Croatia; (S.Š.); (Z.B.)
| | - Zlatko Brkljača
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička 54, 10000 Zagreb, Croatia; (S.Š.); (Z.B.)
| | - Jürgen Kreiter
- Department of Biomedical Sciences, Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, 1210 Vienna, Austria;
| | - Elena E. Pohl
- Department of Biomedical Sciences, Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, 1210 Vienna, Austria;
- Correspondence: (E.E.P.); (M.V.)
| | - Mario Vazdar
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička 54, 10000 Zagreb, Croatia; (S.Š.); (Z.B.)
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, 16610 Prague, Czech Republic
- Correspondence: (E.E.P.); (M.V.)
| |
Collapse
|
35
|
EGLP-1 lowers body weight better than exendin-4 by reducing food intake and increasing basal energy expenditure in diet-induced obese mice. Exp Cell Res 2020; 399:112454. [PMID: 33359447 DOI: 10.1016/j.yexcr.2020.112454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 11/07/2020] [Accepted: 12/18/2020] [Indexed: 12/17/2022]
Abstract
It is well known that GLP-1 activates GLP-1R to reduce body weight by inhibiting eating. GLP-1 is cleaved by the neutral endopeptidase (NEP) 24.11 into a pentapeptide GLP-1 (32-36) amide, which increases basal energy expenditure and inhibits weight gain in obese mice. It is well known that GLP-1 analogs can reduce weight by suppressing eating. However, there are few reports of reducing weight through the dual effects of inhibiting eating and increasing basic energy. Here, we report the peptide EGLP-1, a GLP-1 analogue, which can reduce food intake and increase basal energy expenditure. In C2C12 myotubes, EGLP-1 can increase both phosphorylation of acetyl CoA carboxylase (ACC) and the ratio between phosphorylation of ACC and the total expression of ACC (pACC/ACC). In diet-induced obese mice, EGLP-1 is more effective than exendin-4 in reducing body weight, reducing fat mass and improving hepatic steatosis. At the same time, EGLP-1 can improve hyperglycemia, reduce food intake, and improve insulin resistance, just like exendin-4. In addition, EGLP-1, not exendin-4, can improve physiological parameters associated with lipid metabolism and increase oxygen consumption by increasing uncoupling proteins 3 (UCP3) expression and pACC/ACC ratio in skeletal muscle. Taken together, this data showed that EGLP-1 is able to reduce body weight by reducing food intake and increasing basal energy expenditure, suggesting it may be more effective in treating diabetic and non-diabetic overweight or obese people than pure GLP-1R agonist exendin-4.
Collapse
|
36
|
Thermogenic adipocytes: lineage, function and therapeutic potential. Biochem J 2020; 477:2071-2093. [PMID: 32539124 PMCID: PMC7293110 DOI: 10.1042/bcj20200298] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/13/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022]
Abstract
Metabolic inflexibility, defined as the inability to respond or adapt to metabolic demand, is now recognised as a driving factor behind many pathologies associated with obesity and the metabolic syndrome. Adipose tissue plays a pivotal role in the ability of an organism to sense, adapt to and counteract environmental changes. It provides a buffer in times of nutrient excess, a fuel reserve during starvation and the ability to resist cold-stress through non-shivering thermogenesis. Recent advances in single-cell RNA sequencing combined with lineage tracing, transcriptomic and proteomic analyses have identified novel adipocyte progenitors that give rise to specialised adipocytes with diverse functions, some of which have the potential to be exploited therapeutically. This review will highlight the common and distinct functions of well-known adipocyte populations with respect to their lineage and plasticity, as well as introducing the most recent members of the adipocyte family and their roles in whole organism energy homeostasis. Finally, this article will outline some of the more preliminary findings from large data sets generated by single-cell transcriptomics of mouse and human adipose tissue and their implications for the field, both for discovery and for therapy.
Collapse
|
37
|
Lizcano F, Arroyave F. Control of Adipose Cell Browning and Its Therapeutic Potential. Metabolites 2020; 10:metabo10110471. [PMID: 33227979 PMCID: PMC7699191 DOI: 10.3390/metabo10110471] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/20/2020] [Accepted: 11/02/2020] [Indexed: 12/20/2022] Open
Abstract
Adipose tissue is the largest endocrine organ in humans and has an important influence on many physiological processes throughout life. An increasing number of studies have described the different phenotypic characteristics of fat cells in adults. Perhaps one of the most important properties of fat cells is their ability to adapt to different environmental and nutritional conditions. Hypothalamic neural circuits receive peripheral signals from temperature, physical activity or nutrients and stimulate the metabolism of white fat cells. During this process, changes in lipid inclusion occur, and the number of mitochondria increases, giving these cells functional properties similar to those of brown fat cells. Recently, beige fat cells have been studied for their potential role in the regulation of obesity and insulin resistance. In this context, it is important to understand the embryonic origin of beige adipocytes, the response of adipocyte to environmental changes or modifications within the body and their ability to transdifferentiate to elucidate the roles of these cells for their potential use in therapeutic strategies for obesity and metabolic diseases. In this review, we discuss the origins of the different fat cells and the possible therapeutic properties of beige fat cells.
Collapse
Affiliation(s)
- Fernando Lizcano
- Center of Biomedical Investigation, (CIBUS), Universidad de La Sabana, 250008 Chia, Colombia
- Correspondence:
| | - Felipe Arroyave
- Doctoral Program in Biociencias, Universidad de La Sabana, 250008 Chia, Colombia
| |
Collapse
|
38
|
Cosco J, Scalise M, Colas C, Galluccio M, Martini R, Rovella F, Mazza T, Ecker GF, Indiveri C. ATP modulates SLC7A5 (LAT1) synergistically with cholesterol. Sci Rep 2020; 10:16738. [PMID: 33028978 PMCID: PMC7541457 DOI: 10.1038/s41598-020-73757-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/17/2020] [Indexed: 01/07/2023] Open
Abstract
The plasma membrane transporter hLAT1 is responsible for providing cells with essential amino acids. hLAT1 is over-expressed in virtually all human cancers making the protein a hot-spot in the fields of cancer and pharmacology research. However, regulatory aspects of hLAT1 biology are still poorly understood. A remarkable stimulation of transport activity was observed in the presence of physiological levels of cholesterol together with a selective increase of the affinity for the substrate on the internal site, suggesting a stabilization of the inward open conformation of hLAT1. A synergistic effect by ATP was also observed only in the presence of cholesterol. The same phenomenon was detected with the native protein. Altogether, the biochemical assays suggested that cholesterol and ATP binding sites are close to each other. The computational analysis identified two neighboring regions, one hydrophobic and one hydrophilic, to which cholesterol and ATP were docked, respectively. The computational data predicted interaction of the ϒ-phosphate of ATP with Lys 204, which was confirmed by site-directed mutagenesis. The hLAT1-K204Q mutant showed an impaired function and response to ATP. Interestingly, this residue is conserved in several members of the SLC7 family.
Collapse
Affiliation(s)
- Jessica Cosco
- Department of DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, via Bucci 4C, 87036, Arcavacata di Rende, Italy
| | - Mariafrancesca Scalise
- Department of DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, via Bucci 4C, 87036, Arcavacata di Rende, Italy
| | - Claire Colas
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090, Wien, Austria
| | - Michele Galluccio
- Department of DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, via Bucci 4C, 87036, Arcavacata di Rende, Italy
| | - Riccardo Martini
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090, Wien, Austria
| | - Filomena Rovella
- Department of DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, via Bucci 4C, 87036, Arcavacata di Rende, Italy
| | - Tiziano Mazza
- Department of DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, via Bucci 4C, 87036, Arcavacata di Rende, Italy
| | - Gerhard F Ecker
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse 14, 1090, Wien, Austria
| | - Cesare Indiveri
- Department of DiBEST (Biologia, Ecologia, Scienze Della Terra) Unit of Biochemistry and Molecular Biotechnology, University of Calabria, via Bucci 4C, 87036, Arcavacata di Rende, Italy. .,CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), via Amendola 122/O, 70126, Bari, Italy.
| |
Collapse
|
39
|
Drosophila melanogaster Mitochondrial Carriers: Similarities and Differences with the Human Carriers. Int J Mol Sci 2020; 21:ijms21176052. [PMID: 32842667 PMCID: PMC7504413 DOI: 10.3390/ijms21176052] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/19/2020] [Accepted: 08/19/2020] [Indexed: 12/15/2022] Open
Abstract
Mitochondrial carriers are a family of structurally related proteins responsible for the exchange of metabolites, cofactors and nucleotides between the cytoplasm and mitochondrial matrix. The in silico analysis of the Drosophila melanogaster genome has highlighted the presence of 48 genes encoding putative mitochondrial carriers, but only 20 have been functionally characterized. Despite most Drosophila mitochondrial carrier genes having human homologs and sharing with them 50% or higher sequence identity, D. melanogaster genes display peculiar differences from their human counterparts: (1) in the fruit fly, many genes encode more transcript isoforms or are duplicated, resulting in the presence of numerous subfamilies in the genome; (2) the expression of the energy-producing genes in D. melanogaster is coordinated from a motif known as Nuclear Respiratory Gene (NRG), a palindromic 8-bp sequence; (3) fruit-fly duplicated genes encoding mitochondrial carriers show a testis-biased expression pattern, probably in order to keep a duplicate copy in the genome. Here, we review the main features, biological activities and role in the metabolism of the D. melanogaster mitochondrial carriers characterized to date, highlighting similarities and differences with their human counterparts. Such knowledge is very important for obtaining an integrated view of mitochondrial function in D. melanogaster metabolism.
Collapse
|
40
|
Yuan J, Zhang R, Wu R, Gu Y, Lu Y. The effects of oxytocin to rectify metabolic dysfunction in obese mice are associated with increased thermogenesis. Mol Cell Endocrinol 2020; 514:110903. [PMID: 32531419 DOI: 10.1016/j.mce.2020.110903] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/03/2020] [Accepted: 06/03/2020] [Indexed: 01/12/2023]
Abstract
Oxytocin, a protein hormone mainly produced by hypothalamus, has been shown to repress body weight gain in obese animals, in part, by reducing food intake and increasing energy expenditure. Till now, activation of brown fat tissue (BAT) thermogenesis and white adipose tissue (WAT) browning are considered as two main factors for oxytocin-induced energy expenditure. However, the underlying molecular mechanisms are still not understood well. Here, we observed that oxytocin expression in the hypothalamus and its receptor in adipose tissues were induced by cold exposure in mice. In differentiated adipocytes, oxytocin stimulated brown adipocyte specific gene expression by inducing PRDM16. In high fat diet induced obese mice, oxytocin delivery by osmotic minipumps increased body core temperature and decreased body weight gain. Glucose and insulin tolerance were improved by oxytocin. Hyperinsulinemia and fatty liver were ameliorated in oxytocin-treated animals. Moreover, oxytocin treatment induced thermogenic gene expressions in BAT, inguinal WAT (iWAT), and skeletal muscle. Taken together, our findings revealed a new aspect of oxytocin, i.e. oxytocin induces iWAT browning and stimulates thermogenesis in BAT, iWAT and skeletal muscle, through which oxytocin promotes thermogenesis and thus combats obesity and metabolic dysfunctions.
Collapse
Affiliation(s)
- Jin Yuan
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, Jiangsu, 210011, China; Department of Endocrinology and Metabolism, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Rongping Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Runze Wu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China, 20 Xisi Road, Nantong, Jiangsu, 226001, China
| | - Yunjuan Gu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China, 20 Xisi Road, Nantong, Jiangsu, 226001, China.
| | - Yibing Lu
- Department of Endocrinology, The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing, Jiangsu, 210011, China.
| |
Collapse
|
41
|
Channels and transporters for inorganic ions in plant mitochondria: Prediction and facts. Mitochondrion 2020; 53:224-233. [PMID: 32540403 DOI: 10.1016/j.mito.2020.05.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/01/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023]
Abstract
Mitochondria are crucial bioenergetic organelles for providing different metabolites, including ATP, to sustain cell growth both in animals and in plants. These organelles, delimited by two membranes (outer and inner mitochondrial membrane), maintain their function by an intensive communication with other organelles as well as with the cytosol. Transport of metabolites across the two membranes, but also that of inorganic ions, takes place through specific ion channels and transporters and plays a crucial role in ensuring an adequate ionic milieu within the mitochondria. In the present review we briefly summarize the current knowledge about plant mitochondrial ion channels and transporters in comparison to those of animal mitochondria and examine the possible molecular identity of the so far unidentified transport systems taking into account subcellular targeting predictions and data from literature.
Collapse
|
42
|
Mao X, Huang D, Rao C, Du M, Liang M, Li F, Liu B, Huang K. Enoyl coenzyme A hydratase 1 combats obesity and related metabolic disorders by promoting adipose tissue browning. Am J Physiol Endocrinol Metab 2020; 318:E318-E329. [PMID: 31961704 DOI: 10.1152/ajpendo.00424.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Browning of white adipose tissue (WAT) has been recognized as an important strategy for the treatment of obesity, insulin resistance, and diabetes. Enoyl coenzyme A hydratase 1 (ECH1) is a widely known enzyme involved in lipid metabolism. However, whether and how ECH1 is implicated in browning of WAT remain obscure. Adeno-associated, virus-mediated genetic engineering of ECH1 in adipose tissue was used in investigations in mouse models of obesity induced by a high-fat diet (HFD) or browning induced by cold exposure. Metabolic parameters showed that ECH1 overexpression decreased weight gain and improved insulin sensitivity and lipid profile after 8 wk of an HFD. Further work revealed that these changes were associated with enhanced energy expenditure and increased appearance of brown-like adipocytes in inguinal WAT, as verified by a remarkable increase in uncoupling protein 1 and thermogenic gene expression. In vitro, ECH1 induced brown fat-related gene expression in adipocytes differentiated from primary stromal vascular fractions, whereas knockdown of ECH1 reversed this effect. Mechanistically, ECH1 regulated the thermogenic program by inhibiting mammalian target of rapamycin signaling, which may partially explain the potential mechanism for ECH1 regulating adipose browning. In summary, ECH1 may participate in the pathology of obesity by regulating browning of WAT, which probably provides us with a new therapeutic strategy for combating obesity.
Collapse
Affiliation(s)
- Xiaoxiang Mao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dandan Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Caijun Rao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Du
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minglu Liang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Baoqing Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
43
|
Dhamrait GK, Panchal K, Fleury NJ, Abel TN, Ancliffe MK, Crew RC, Croft K, Fernandez BO, Minnion M, Hart PH, Lucas RM, Mark PJ, Feelisch M, Weller RB, Matthews V, Gorman S. Characterising nitric oxide-mediated metabolic benefits of low-dose ultraviolet radiation in the mouse: a focus on brown adipose tissue. Diabetologia 2020; 63:179-193. [PMID: 31713010 DOI: 10.1007/s00125-019-05022-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 08/29/2019] [Indexed: 12/20/2022]
Abstract
AIMS/HYPOTHESIS Exposure to sunlight has the potential to suppress metabolic dysfunction and obesity. We previously demonstrated that regular exposure to low-doses of ultraviolet radiation (UVR) reduced weight gain and signs of diabetes in male mice fed a high-fat diet, in part via release of nitric oxide from skin. Here, we explore further mechanistic pathways through which low-dose UVR exerts these beneficial effects. METHODS We fed mice with a luciferase-tagged Ucp1 gene (which encodes uncoupling protein-1 [UCP-1]), referred to here as the Ucp1 luciferase transgenic mouse ('Thermomouse') a high-fat diet and examined the effects of repeated exposure to low-dose UVR on weight gain and development of metabolic dysfunction as well as UCP-1-dependent thermogenesis in interscapular brown adipose tissue (iBAT). RESULTS Repeated exposure to low-dose UVR suppressed the development of glucose intolerance and hepatic lipid accumulation via dermal release of nitric oxide while also reducing circulating IL-6 (compared with mice fed a high-fat diet only). Dietary nitrate supplementation did not mimic the effects of low-dose UVR. A single low dose of UVR increased UCP-1 expression (by more than twofold) in iBAT of mice fed a low-fat diet, 24 h after exposure. However, in mice fed a high-fat diet, there was no effect of UVR on UCP-1 expression in iBAT (compared with mock-treated mice) when measured at regular intervals over 12 weeks. More extensive circadian studies did not identify any substantial shifts in UCP-1 expression in mice exposed to low-dose UVR, although skin temperature at the interscapular site was reduced in UVR-exposed mice. The appearance of cells with a white adipocyte phenotype ('whitening') in iBAT induced by consuming the high-fat diet was suppressed by exposure to low-dose UVR in a nitric oxide-dependent fashion. Significant shifts in the expression of important core gene regulators of BAT function (Dio2, increased more than twofold), fatty acid transport (increased Fatp2 [also known as Slc27a2]), lipolysis (decreased Atgl [also known as Pnpla2]), lipogenesis (decreased Fasn) and inflammation (decreased Tnf), and proportions of macrophages (increased twofold) were observed in iBAT of mice exposed to low-dose UVR. These effects were independent of nitric oxide released from skin. CONCLUSIONS/INTERPRETATION Our results suggest that non-burning (low-dose) UVR suppresses the BAT 'whitening', steatotic and pro-diabetic effects of consuming a high-fat diet through skin release of nitric oxide, with some metabolic and immune pathways in iBAT regulated by UVR independently of nitric oxide.
Collapse
Affiliation(s)
- Gursimran K Dhamrait
- Telethon Kids Institute, University of Western Australia, PO Box 855, West Perth, WA, 6782, Australia
| | - Kunjal Panchal
- Telethon Kids Institute, University of Western Australia, PO Box 855, West Perth, WA, 6782, Australia
| | - Naomi J Fleury
- Telethon Kids Institute, University of Western Australia, PO Box 855, West Perth, WA, 6782, Australia
| | - Tamara N Abel
- Telethon Kids Institute, University of Western Australia, PO Box 855, West Perth, WA, 6782, Australia
| | - Mathew K Ancliffe
- Telethon Kids Institute, University of Western Australia, PO Box 855, West Perth, WA, 6782, Australia
| | - Rachael C Crew
- School of Human Sciences, University of Western Australia, Perth, WA, Australia
| | - Kevin Croft
- School of Biomedical Science - Royal Perth Hospital Unit, The University of Western Australia, Perth, WA, Australia
| | - Bernadette O Fernandez
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Magdalena Minnion
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Prue H Hart
- Telethon Kids Institute, University of Western Australia, PO Box 855, West Perth, WA, 6782, Australia
| | - Robyn M Lucas
- National Centre for Epidemiology and Population Health, Research School of Population Health, Australian National University, Canberra, ACT, Australia
- Centre for Ophthalmology and Visual Science, University of Western Australia, Perth, WA, Australia
| | - Peter J Mark
- School of Human Sciences, University of Western Australia, Perth, WA, Australia
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Richard B Weller
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Vance Matthews
- School of Biomedical Science - Royal Perth Hospital Unit, The University of Western Australia, Perth, WA, Australia
| | - Shelley Gorman
- Telethon Kids Institute, University of Western Australia, PO Box 855, West Perth, WA, 6782, Australia.
| |
Collapse
|
44
|
Chen KY, Brychta RJ, Abdul Sater Z, Cassimatis TM, Cero C, Fletcher LA, Israni NS, Johnson JW, Lea HJ, Linderman JD, O'Mara AE, Zhu KY, Cypess AM. Opportunities and challenges in the therapeutic activation of human energy expenditure and thermogenesis to manage obesity. J Biol Chem 2019; 295:1926-1942. [PMID: 31914415 DOI: 10.1074/jbc.rev119.007363] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The current obesity pandemic results from a physiological imbalance in which energy intake chronically exceeds energy expenditure (EE), and prevention and treatment strategies remain generally ineffective. Approaches designed to increase EE have been informed by decades of experiments in rodent models designed to stimulate adaptive thermogenesis, a long-term increase in metabolism, primarily induced by chronic cold exposure. At the cellular level, thermogenesis is achieved through increased rates of futile cycling, which are observed in several systems, most notably the regulated uncoupling of oxidative phosphorylation from ATP generation by uncoupling protein 1, a tissue-specific protein present in mitochondria of brown adipose tissue (BAT). Physiological activation of BAT and other organ thermogenesis occurs through β-adrenergic receptors (AR), and considerable effort over the past 5 decades has been directed toward developing AR agonists capable of safely achieving a net negative energy balance while avoiding unwanted cardiovascular side effects. Recent discoveries of other BAT futile cycles based on creatine and succinate have provided additional targets. Complicating the current and developing pharmacological-, cold-, and exercise-based methods to increase EE is the emerging evidence for strong physiological drives toward restoring lost weight over the long term. Future studies will need to address technical challenges such as how to accurately measure individual tissue thermogenesis in humans; how to safely activate BAT and other organ thermogenesis; and how to sustain a negative energy balance over many years of treatment.
Collapse
Affiliation(s)
- Kong Y Chen
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892.
| | - Robert J Brychta
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Zahraa Abdul Sater
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Thomas M Cassimatis
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Cheryl Cero
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Laura A Fletcher
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Nikita S Israni
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - James W Johnson
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Hannah J Lea
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Joyce D Linderman
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Alana E O'Mara
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Kenneth Y Zhu
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, NIDDK, National Institutes of Health, Bethesda, Maryland 20892.
| |
Collapse
|
45
|
Ostrakhovitch EA, Akakura S, Sanokawa-Akakura R, Tabibzadeh S. 3-Mercaptopyruvate sulfurtransferase disruption in dermal fibroblasts facilitates adipogenic trans-differentiation. Exp Cell Res 2019; 385:111683. [DOI: 10.1016/j.yexcr.2019.111683] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 10/13/2019] [Accepted: 10/17/2019] [Indexed: 12/17/2022]
|
46
|
Ng SP, Nomura W, Mohri S, Takahashi H, Jheng HF, Ara T, Nagai H, Ito T, Kawada T, Goto T. Soy hydrolysate enhances the isoproterenol-stimulated lipolytic pathway through an increase in β-adrenergic receptor expression in adipocytes. Biosci Biotechnol Biochem 2019; 83:1782-1789. [PMID: 31045477 DOI: 10.1080/09168451.2019.1611413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/13/2019] [Indexed: 01/20/2023]
Abstract
Activation of the adipose lipolytic pathway during lipid metabolism is mediated by protein kinase A (PKA), which responds to β-adrenergic stimulation, leading to increased lipolysis. Soy is well known as a functional food and it is able to affect lipolysis in adipocytes. However, the mechanism by which soy components contribute to the lipolytic pathway remains to be fully elucidated. Here, we show that hydrolyzed soy enhances isoproterenol-stimulated lipolysis and activation of PKA in 3T3-L1 adipocytes. We also found that the expression of β-adrenergic receptors, which coordinate the activation of PKA, is elevated in adipocytes differentiated in the presence of soy hydrolysate. The activity of the soy hydrolysate towards β-adrenergic receptor expression was detected in its hydrophilic fraction. Our results suggest that the soy hydrolysate enhances the PKA pathway through the upregulation of β-adrenergic receptor expression and thereby, increase lipolysis in adipocytes.
Collapse
Affiliation(s)
- Su-Ping Ng
- a Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University , Uji , Japan
| | - Wataru Nomura
- a Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University , Uji , Japan
- b Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University , Kyoto , Japan
| | - Shinsuke Mohri
- a Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University , Uji , Japan
| | - Haruya Takahashi
- a Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University , Uji , Japan
| | - Huei-Fen Jheng
- a Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University , Uji , Japan
| | - Takeshi Ara
- a Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University , Uji , Japan
| | - Hiroyuki Nagai
- c Gifu Prefectural Research Institute for Health and Environmental Sciences , Kakamigahara , Japan
| | - Tetsuro Ito
- c Gifu Prefectural Research Institute for Health and Environmental Sciences , Kakamigahara , Japan
| | - Teruo Kawada
- a Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University , Uji , Japan
- b Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University , Kyoto , Japan
| | - Tsuyoshi Goto
- a Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University , Uji , Japan
- b Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University , Kyoto , Japan
| |
Collapse
|
47
|
Mäkelä AM, Hohtola E, Miinalainen IJ, Autio JA, Schmitz W, Niemi KJ, Hiltunen JK, Autio KJ. Mitochondrial 2,4-dienoyl-CoA reductase (Decr) deficiency and impairment of thermogenesis in mouse brown adipose tissue. Sci Rep 2019; 9:12038. [PMID: 31427678 PMCID: PMC6700156 DOI: 10.1038/s41598-019-48562-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 08/07/2019] [Indexed: 12/17/2022] Open
Abstract
A large number of studies have demonstrated significance of polyunsaturated fatty acids (PUFAs) for human health. However, many aspects on signals translating PUFA-sensing into body homeostasis have remained enigmatic. To shed light on PUFA physiology, we have generated a mouse line defective in mitochondrial dienoyl-CoA reductase (Decr), which is a key enzyme required for β-oxidation of PUFAs. Previously, we have shown that these mice, whose oxidation of saturated fatty acid is intact but break-down of unsaturated fatty acids is blunted, develop severe hypoglycemia during metabolic stresses and fatal hypothermia upon acute cold challenge. In the current work, indirect calorimetry and thermography suggested that cold intolerance of Decr−/− mice is due to failure in maintaining appropriate heat production at least partly due to failure of brown adipose tissue (BAT) thermogenesis. Magnetic resonance imaging, electron microscopy, mass spectrometry and biochemical analysis showed attenuation in activation of lipolysis despite of functional NE-signaling and inappropriate expression of genes contributing to thermogenesis in iBAT when the Decr−/− mice were exposed to cold. We hypothesize that the failure in turning on BAT thermogenesis occurs due to accumulation of unsaturated long-chain fatty acids or their metabolites in Decr−/− mice BAT suppressing down-stream propagation of NE-signaling.
Collapse
Affiliation(s)
- Anne M Mäkelä
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Esa Hohtola
- Department of Ecology and Genetics, University of Oulu, Oulu, Finland
| | | | - Joonas A Autio
- Center for Biosystems Dynamics Research, RIKEN, Kobe, Japan.,Medical Research Center, University of Oulu and Oulu University Hospital, Oulu, Finland
| | | | - Kalle J Niemi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - J Kalervo Hiltunen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Kaija J Autio
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
| |
Collapse
|
48
|
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: 170] [Impact Index Per Article: 28.3] [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
|
49
|
Abstract
Uncoupling protein 1 (UCP1) is an integral protein of the inner mitochondrial membrane (IMM) that is expressed specifically in brown and beige fat depots. UCP1 is responsible for the production of heat to control core body temperature, the regulation of fat metabolism, and the energy balance. As an uncoupling protein, UCP1 transports H+ across the IMM in presence of long-chain fatty acids (FA), which makes brown fat mitochondria produce heat at the expense of ATP. However, the exact mechanism of UCP1 action has remained difficult to elucidate, because direct methods for studying currents generated by UCP1 were unavailable. Recently, the patch-clamp technique was successfully applied to brown and beige fat mitochondria to directly study H+ currents across the IMM and characterize UCP1 function. A new model of the UCP1 mechanism was proposed based on the patch-clamp analysis. In this model, both FA anions (FA-) and H+ are transport substrates of UCP1, and UCP1 operates as a non-canonical FA-/H+ symporter. Here, we summarize recent findings obtained with the patch-clamp technique that describe how UCP1 can transport not only H+ but also FA-.
Collapse
|
50
|
Adipose lipidomics and RNA-Seq analysis revealed the enhanced mitochondrial function in UCP1 knock-in pigs. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1375-1383. [PMID: 31271850 DOI: 10.1016/j.bbalip.2019.06.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 05/31/2019] [Accepted: 06/28/2019] [Indexed: 11/24/2022]
Abstract
Uncoupling protein 1 (UCP1) plays a key role in nonshivering thermogenesis and is involved in the pathogenesis of obesity. In a previous study, we generated adipocyte-specific UCP1 knock-in (UCP1-KI) pigs, which exhibited improved thermoregulatory ability and decreased fat deposition. To investigate whether UCP1 knock-in alters the lipid composition of adipose tissues, lipidomics of inguinal subcutaneous white adipose tissue (iWAT) and backfat from 6-month-old cold-treated UCP1-KI pigs and wild-type (WT) pigs were profiled. In addition, genome-wide RNA-sequencing of iWAT was performed to further study the genetic basis for lipid alterations. The results showed that iWAT and backfat from UCP1-KI pigs exhibited distinct lipidomic profiles, as the mild lipid alteration was observed in backfat of UCP1 knock-in pigs. Inguinal WAT from UCP1-KI pigs contained significantly decreased total triacylglycerol (p < 0.05), together with the downregulation of genes involved in fatty acid metabolism, suggesting the decreased lipogenesis in iWAT of UCP1-KI pigs. Significantly increased levels of total sphingolipids (p<0.05) were also observed in iWAT from UCP1-KI pigs. Notably, two mitochondrial-specific lipid species, cardiolipin CL72:8 (18:2) and CL74:9 (18:2), were found to be dramatically increased in iWAT from UCP1-KI pigs, suggesting enhanced mitochondrial function. This observation was further supported by the significant upregulation of numerous mitochondrial-related genes and significantly increased number of large mitochondria and mitochondrial cristae in iWAT of UCP1-KI pigs. Taken together, these data illustrate the specific role of UCP1 in lipid metabolism of fat tissues in pigs and provide new data for characterization of fat traits in UCP1-KI pigs.
Collapse
|