1
|
Abdillah AM, Lee JY, Lee YR, Yun JW. Modulatory roles of capsaicin on thermogenesis in C2C12 myoblasts and the skeletal muscle of mice. Chem Biol Interact 2025; 407:111380. [PMID: 39800145 DOI: 10.1016/j.cbi.2025.111380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/25/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Capsaicin, a polyphenol, is known to regulate energy expenditure and thermogenesis in adipocytes and muscles. However, its role in modulating uncoupling proteins (UCPs) and adenosine triphosphate (ATP)-dependent thermogenesis in muscles remains unclear. This study investigated the mechanisms underlying the role of capsaicin in modulating the UCP- and ATP-dependent thermogenesis in C2C12 myoblasts, as well as the gastrocnemius (GM) and soleus muscles (SM) of mice. We employed molecular dynamics (MD), quantitative real-time polymerase chain reactions (qRT-PCR), immunoblots, staining methods, and assay kits to investigate the role of capsaicin on thermogenesis and its modulatory roles on the transient receptor potential cation channel subfamily V member 1 (TRPV1) and α-/β-adrenergic receptors (ARs) using in vitro and in vivo models. Our findings demonstrate that capsaicin treatment in high-fat diet-induced obese mice reduces weight gain and elevates the expression of UCP- and ATP-dependent thermogenic effectors through ATP-consuming calcium and creatine futile cycles. In vitro and in vivo models capsaicin treatment elevated the expression of sarcoendoplasmic/endoplasmic reticulum calcium ATPases (SERCA-1 and -2), ryanodine receptors (RYR-1 and -2), uncoupling proteins (UCP-2 and -3), creatine kinase B (CKB), and creatine kinase mitochondrial 2 (CKMT2), through activation of TRPV1, α1-, β2-, and β3-AR as well as the suppressed expression of α2-AR. Furthermore, our results also indicate that capsaicin promotes myotube development and enhances lipid metabolism in C2C12 cells. We found that capsaicin increased intracellular Ca2+ levels and the expression of the voltage-dependent anion channel (VDAC) and mitochondrial calcium uniporter (MCU), suggesting that elevated mitochondrial Ca2+ levels boost the expression of oxidative phosphorylation protein complexes via the activation of the ATP-futile cycle. Mechanistic studies in C2C12 cells revealed that TRPV1 is likely dispensable for capsaicin-induced thermogenesis, and TRPV1 and α1-AR may synergistically induce thermogenesis. Collectively, our findings have uncovered a novel mechanism of UCP- and ATP-dependent thermogenesis and its associated pathways in both cellular and animal models which is crucial for designing therapeutic strategies to address obesity and associated metabolic diseases.
Collapse
Affiliation(s)
- Alfin Mohammad Abdillah
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea
| | - Jae Young Lee
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea
| | - Young Rok Lee
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea
| | - Jong Won Yun
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea.
| |
Collapse
|
2
|
Cypess AM, Cannon B, Nedergaard J, Kazak L, Chang DC, Krakoff J, Tseng YH, Schéele C, Boucher J, Petrovic N, Blondin DP, Carpentier AC, Virtanen KA, Kooijman S, Rensen PCN, Cero C, Kajimura S. Emerging debates and resolutions in brown adipose tissue research. Cell Metab 2025; 37:12-33. [PMID: 39644896 PMCID: PMC11710994 DOI: 10.1016/j.cmet.2024.11.002] [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: 03/26/2024] [Revised: 08/28/2024] [Accepted: 11/01/2024] [Indexed: 12/09/2024]
Abstract
Until two decades ago, brown adipose tissue (BAT) was studied primarily as a thermogenic organ of small rodents in the context of cold adaptation. The discovery of functional human BAT has opened new opportunities to understand its physiological role in energy balance and therapeutic applications for metabolic disorders. Significantly, the role of BAT extends far beyond thermogenesis, including glucose and lipid homeostasis, by releasing mediators that communicate with other cells and organs. The field has made major advances by using new model systems, ranging from subcellular studies to clinical trials, which have also led to debates. In this perspective, we identify six fundamental issues that are currently controversial and comprise dichotomous models. Each side presents supporting evidence and, critically, the necessary methods and falsifiable experiments that would resolve the dispute. With this collaborative approach, the field will continue to productively advance the understanding of BAT physiology, appreciate the importance of thermogenic adipocytes as a central area of ongoing research, and realize the therapeutic potential.
Collapse
Affiliation(s)
- 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.
| | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Lawrence Kazak
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Douglas C Chang
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ 85016, USA
| | - Jonathan Krakoff
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ 85016, USA
| | - Yu-Hua Tseng
- Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA
| | - Camilla Schéele
- Novo Nordisk Foundation Center for Basic Metabolic Research, The Center of Inflammation and Metabolism and the Center for Physical Activity Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Natasa Petrovic
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91 Stockholm, Sweden
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, the Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, the Netherlands
| | - Cheryl Cero
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
3
|
Qiu J, Guo Y, Guo X, Liu Z, Li Z, Zhang J, Cao Y, Li J, Yu S, Xu S, Chen J, Wang D, Yu J, Guo M, Zhou W, Wang S, Wang Y, Ma X, Xie C, Xu L. Ucp1 Ablation Improves Skeletal Muscle Glycolytic Function in Aging Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411015. [PMID: 39569747 PMCID: PMC11727132 DOI: 10.1002/advs.202411015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/06/2024] [Indexed: 11/22/2024]
Abstract
Muscular atrophy is among the systematic decline in organ functions in aging, while defective thermogenic fat functionality precedes these anomalies. The potential crosstalk between adipose tissue and muscle during aging is poorly understood. In this study, it is showed that UCP1 knockout (KO) mice characterized deteriorated brown adipose tissue (BAT) function in aging, yet their glucose homeostasis is sustained and energy expenditure is increased, possibly compensated by improved inguinal adipose tissue (iWAT) and muscle functionality compared to age-matched WT mice. To understand the potential crosstalk, RNA-seq and metabolomic analysis were performed on adipose tissue and muscle in aging mice and revealed that creatine levels are increased both in iWAT and muscle of UCP1 KO mice. Interestingly, molecular analysis and metabolite tracing revealed that creatine biosynthesis is increased in iWAT while creatine uptake is increased in muscle in UCP1 KO mice, suggesting creatine transportation from iWAT to muscle. Importantly, creatine analog β-GPA abolished the differences in muscle functions between aging WT and UCP1 KO mice, while UCP1 inhibitor α-CD improved muscle glycolytic function and glucose metabolism in aging mice. Overall, these results suggested that iWAT and skeletal muscle compensate for declined BAT function during aging via creatine metabolism to sustain metabolic homeostasis.
Collapse
Affiliation(s)
- Jin Qiu
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Yuhan Guo
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Xiaozhen Guo
- State Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Ziqi Liu
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Zixuan Li
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Jun Zhang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Yutang Cao
- School of Chinese Materia MedicaNanjing University of Chinese MedicineNanjing210023China
| | - Jiaqi Li
- State Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- University of Chinese Academy of SciencesBeijing100049China
| | - Shuwu Yu
- State Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- University of Chinese Academy of SciencesBeijing100049China
| | - Sainan Xu
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Juntong Chen
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Dongmei Wang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Jian Yu
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
- Department of Endocrinology and MetabolismFengxian Central Hospital Affiliated to Southern Medical UniversityShanghai201499China
| | - Mingwei Guo
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Wenhao Zhou
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Sainan Wang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Yiwen Wang
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| | - Xinran Ma
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
- Shanghai Frontiers Science Center of Genome Editing and Cell TherapyShanghai Key Laboratory of Regulatory Biology and School of Life SciencesEast China Normal UniversityShanghai200241China
- Chongqing Key Laboratory of Precision OpticsChongqing Institute of East China Normal UniversityChongqing401120China
| | - Cen Xie
- State Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- School of Chinese Materia MedicaNanjing University of Chinese MedicineNanjing210023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory BiologyInstitute of Biomedical Sciences and School of Life SciencesEast China Normal UniversityShanghai200241China
| |
Collapse
|
4
|
Roth L, Hoffmann A, Hagemann T, Wagner L, Strehlau C, Sheikh B, Donndorf L, Ghosh A, Noé F, Wolfrum C, Krohn K, Weiner J, Heiker JT, Klöting N, Stumvoll M, Tönjes A, Blüher M, Mittag J, Krause K. Thyroid hormones are required for thermogenesis of beige adipocytes induced by Zfp423 inactivation. Cell Rep 2024; 43:114987. [PMID: 39580797 DOI: 10.1016/j.celrep.2024.114987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/30/2024] [Accepted: 10/31/2024] [Indexed: 11/26/2024] Open
Abstract
The significance of thyroid hormones (THs) in beige adipocyte thermogenesis remains incompletely understood. We previously reported that THs directly regulate the expression of zinc-finger protein 423 (ZFP423), an anti-thermogenic factor, in adipose tissue. This study investigates the interaction between THs and adrenergic signaling in regulating thermogenic capacity and activation of beige adipocytes formed in response to Zfp423 deletion. We demonstrate that THs are indispensable for uncoupling protein 1 (UCP1)-dependent thermogenesis, leading to increased energy expenditure in mice with adipocyte-specific Zfp423 knockout. Targeted activation of the thyroid receptor isoform TRβ, which plays a central role in the inguinal depot, is sufficient to enhance energy expenditure in hypothyroid Zfp423iAKO mice. Mechanistically, THs and ZFP423 pathways cooperate to regulate early B cell factor 2 (EBF2)-mediated activation of the Ucp1 gene. RNA sequencing (RNA-seq) analysis of human adipose tissue samples supports the relevance of this regulatory network for human adipose tissue plasticity.
Collapse
Affiliation(s)
- Lisa Roth
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Tobias Hagemann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Leonie Wagner
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Christian Strehlau
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Bilal Sheikh
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Lorenz Donndorf
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Adhideb Ghosh
- Institute of Food, Nutrition and Health, ETH Zurich, 8092 Zürich, Switzerland
| | - Falko Noé
- Institute of Food, Nutrition and Health, ETH Zurich, 8092 Zürich, Switzerland
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zurich, 8092 Zürich, Switzerland
| | - Knut Krohn
- Medical Faculty, Center for DNA Technologies, University of Leipzig, 04103 Leipzig, Germany
| | - Juliane Weiner
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - John T Heiker
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Michael Stumvoll
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Anke Tönjes
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany
| | - Matthias Blüher
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München, University of Leipzig and University Hospital Leipzig, 04103 Leipzig, Germany
| | - Jens Mittag
- Institute of Experimental Endocrinology/CBBM, University of Lübeck, 23562 Lübeck, Germany
| | - Kerstin Krause
- Department of Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; German Center for Diabetes Research e.V., 85764 Neuherberg, Germany.
| |
Collapse
|
5
|
Renzi G, Vlassakev I, Hansen M, Higos R, Lecoutre S, Elmastas M, Hodek O, Moritz T, Alaeddine LM, Frendo-Cumbo S, Dahlman I, Kerr A, Maqdasy S, Mejhert N, Rydén M. Epigenetic suppression of creatine kinase B in adipocytes links endoplasmic reticulum stress to obesity-associated inflammation. Mol Metab 2024; 92:102082. [PMID: 39675471 PMCID: PMC11731883 DOI: 10.1016/j.molmet.2024.102082] [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: 10/25/2024] [Revised: 11/26/2024] [Accepted: 12/05/2024] [Indexed: 12/17/2024] Open
Abstract
In white adipose tissue, disturbed creatine metabolism through reduced creatine kinase B (CKB) transcription contributes to obesity-related inflammation. However, the mechanisms regulating CKB expression in human white adipocytes remain unclear. By screening conditions perturbed in obesity, we identified endoplasmic reticulum (ER) stress as a key suppressor of CKB transcription across multiple cell types. Through follow-up studies, we found that ER stress through the IRE1-XBP1s pathway, promotes CKB promoter methylation via the methyltransferase DNMT3A. This epigenetic change represses CKB transcription, shifting metabolism towards glycolysis and increasing the production of the pro-inflammatory chemokine CCL2. We validated our findings in vivo, demonstrating that individuals living with obesity show an inverse relationship between CKB expression and promoter methylation in white adipocytes, along with elevated CCL2 secretion. Overall, our study uncovers a regulatory axis where ER stress drives inflammation in obesity by reducing CKB abundance, and consequently altering the bioenergetic state of the cell.
Collapse
Affiliation(s)
- Gianluca Renzi
- Department of Medicine (H7), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, SE-141 83, Huddinge, Sweden
| | - Ivan Vlassakev
- Department of Medicine (H7), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, SE-141 83, Huddinge, Sweden
| | - Mattias Hansen
- Department of Medicine (H7), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, SE-141 83, Huddinge, Sweden
| | - Romane Higos
- Department of Medicine (H7), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, SE-141 83, Huddinge, Sweden
| | - Simon Lecoutre
- Nutrition and Obesities: Systemic Approaches Research Group (Nutri-Omics), Sorbonne Université, INSERM, F-75013 Paris, France
| | - Merve Elmastas
- Department of Medicine (H7), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, SE-141 83, Huddinge, Sweden
| | - Ondrej Hodek
- Swedish Metabolomics Center, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden
| | - Thomas Moritz
- Swedish Metabolomics Center, Department of Forest Genetics and Plant Physiology, Swedish University of Agricultural Sciences, Umeå, Sweden; The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lynn M Alaeddine
- Department of Medicine (H7), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, SE-141 83, Huddinge, Sweden
| | - Scott Frendo-Cumbo
- Department of Medicine (H7), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, SE-141 83, Huddinge, Sweden
| | - Ingrid Dahlman
- Department of Clinical Science and Education, Karolinska Institutet, Stockholm, Sweden
| | - Alastair Kerr
- Department of Medicine (H7), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, SE-141 83, Huddinge, Sweden
| | - Salwan Maqdasy
- Department of Medicine (H7), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, SE-141 83, Huddinge, Sweden
| | - Niklas Mejhert
- Department of Medicine (H7), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, SE-141 83, Huddinge, Sweden; Steno Diabetes Center, Copenhagen, Herlev, Denmark.
| | - Mikael Rydén
- Department of Medicine (H7), Karolinska Institutet, ME Endokrinologi, Karolinska University Hospital Huddinge, SE-141 83, Huddinge, Sweden; Steno Diabetes Center, Copenhagen, Herlev, Denmark.
| |
Collapse
|
6
|
Geng Y, DeLay SL, Chen X, Miska J. It Is Not Just About Storing Energy: The Multifaceted Role of Creatine Metabolism on Cancer Biology and Immunology. Int J Mol Sci 2024; 25:13273. [PMID: 39769038 PMCID: PMC11678534 DOI: 10.3390/ijms252413273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Creatine, a naturally occurring compound in mammals, is crucial in energy metabolism, particularly within muscle and brain tissues. While creatine metabolism in cancer has been studied for several decades, emerging studies are beginning to clarify the sometimes-contradictory role creatine has in either the promotion or inhibition of cancer. On one hand, creatine can directly enhance anti-tumor CD8+ T-cell activity and induce tumor apoptosis, contributing to antitumor immunity. Conversely, other studies have shown that creatine can facilitate cancer cell growth and migration by providing an energy source and activating several signaling pathways. This review will examine what is known about creatine in cancer biology, with a focus on understanding its roles across different cellular compartments. Lastly, we discuss the emerging roles of creatine metabolism, providing exciting new insights into this often-overlooked pathway. This review highlights the complex role of creatine in cancer development and treatment, offering insights into its potential as both a therapeutic target and a risk factor in oncogenesis.
Collapse
Affiliation(s)
- Yuheng Geng
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Susan L. DeLay
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Xiaoyang Chen
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Jason Miska
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
7
|
Chang L, Meng F, Jiao B, Zhou T, Su R, Zhu C, Wu Y, Ling Y, Wang S, Wu K, Zhang D, Cao J. Integrated analysis of omics reveals the role of scapular fat in thermogenesis adaptation in sunite sheep. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101292. [PMID: 39018792 DOI: 10.1016/j.cbd.2024.101292] [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: 04/23/2024] [Revised: 06/30/2024] [Accepted: 07/11/2024] [Indexed: 07/19/2024]
Abstract
Inhabiting some of the world's most inhospitable climatic regions, the Sunite Mongolian sheep generates average temperatures as low as 4.3 °C and a minimum temperature of -38.8 °C; in these environments, they make essential cold adaptations. In this regard, scapular fat tissues from Mongolian sheep were collected both in winter and summer for transcriptomic and proteomic analyses to identify genes related to adaptive thermogenesis. In the transcriptome analysis, 588 differentially expressed genes were identified to participate in smooth muscle activity and fat metabolism, as well as in nutrient regulation. There were 343 upregulated and 245 downregulated genes. GO and KEGG pathway analyses on these genes revealed their participation in regulating smooth muscle activity, metabolism of fats, and nutrients. Proteomic analysis showed the differential expression of 925 proteins: among them, there are 432 up- and 493 down-expressed proteins. These proteins are mainly involved in oxidative phosphorylation, respiratory chain complex assembly, and ATP production by electron transport. Furthermore, using both sets at a more detailed level of analysis revealed over-representation in gene ontology categories related to hormone signaling, metabolism of lipids, the pentose phosphate pathway, the TCA cycle, and especially the process of oxidative phosphorylation. The identified essential genes and proteins were further validated by quantitative real-time polymerase chain reaction and Western blotting, respectively; key metabolic network constriction was constructed. The present study emphasized the critical role of lipid turnover in scapular fat for thermogenic adaptation in Sunite sheep.
Collapse
Affiliation(s)
- Longwei Chang
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China
| | - Fanhua Meng
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China.
| | - Boran Jiao
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China
| | - Tong Zhou
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China
| | - Rina Su
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China
| | - Chunxiao Zhu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China
| | - Yi Wu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China
| | - Yu Ling
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China
| | - Shenyuan Wang
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China
| | - Kaifeng Wu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China
| | - Dong Zhang
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China.
| | - Junwei Cao
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China.
| |
Collapse
|
8
|
Berriel Diaz M, Rohm M, Herzig S. Cancer cachexia: multilevel metabolic dysfunction. Nat Metab 2024; 6:2222-2245. [PMID: 39578650 DOI: 10.1038/s42255-024-01167-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 10/16/2024] [Indexed: 11/24/2024]
Abstract
Cancer cachexia is a complex metabolic disorder marked by unintentional body weight loss or 'wasting' of body mass, driven by multiple aetiological factors operating at various levels. It is associated with many malignancies and significantly contributes to cancer-related morbidity and mortality. With emerging recognition of cancer as a systemic disease, there is increasing awareness that understanding and treatment of cancer cachexia may represent a crucial cornerstone for improved management of cancer. Here, we describe the metabolic changes contributing to body wasting in cachexia and explain how the entangled action of both tumour-derived and host-amplified processes induces these metabolic changes. We discuss energy homeostasis and possible ways that the presence of a tumour interferes with or hijacks physiological energy conservation pathways. In that context, we highlight the role played by metabolic cross-talk mechanisms in cachexia pathogenesis. Lastly, we elaborate on the challenges and opportunities in the treatment of this devastating paraneoplastic phenomenon that arise from the complex and multifaceted metabolic cross-talk mechanisms and provide a status on current and emerging therapeutic approaches.
Collapse
Affiliation(s)
- Mauricio Berriel Diaz
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Chair Molecular Metabolic Control, Technical University of Munich, Munich, Germany.
| |
Collapse
|
9
|
Ren Y, Huang P, Zhang L, Tang Y, He S, Li H, Huang X, Ding Y, Liu L, Liu L, He X. Multi-omics landscape of childhood simple obesity: novel insights into pathogenesis and biomarkers discovery. Cell Biosci 2024; 14:145. [PMID: 39609876 PMCID: PMC11606102 DOI: 10.1186/s13578-024-01322-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND The increasing incidence of childhood obesity annually has led to a surge in physical and mental health risks, making it a significant global public health concern. This study aimed to discover novel biomarkers of childhood simple obesity through integrative multi-omics analysis, uncovering their potential connections and providing fresh research directions for the complex pathogenesis and treatment strategies. METHODS Transcriptome, untargeted metabolome, and 16 S rDNA sequencing were conducted on subjects to examine transcripts, metabolites in blood, and gut microflora in stool. RESULTS Transcriptomic analysis identified 599 differentially expressed genes (DEGs), of which 25 were immune-related genes, and participated in immune pathways such as antimicrobial peptides, neutrophil degranulation, and interferons. The optimal random forest model based on these genes exhibited an AUC of 0.844. The metabolomic analysis examined 71 differentially expressed metabolites (DEMs), including 12 immune-related metabolites. Notably, lauric acid showed an extremely strong positive correlation with BMI and showed a good discriminative power for obesity (AUC = 0.82). DEMs were found to be significantly enriched in four metabolic pathways, namely "Aminoacyl-tRNA biosynthesis", "Valine leucine and isoleucine biosynthesis, and Glycine", "Serine and threonine metabolism", and "Biosynthesis of unsaturated fatty acids". Microbiome analysis revealed 12 differential gut microbiotas (DGMs) at the phylum and genus levels, with p_Firmicutes dominating in the obese group and g_Escherichia-Shigella in the normal group. Subsequently, a Random Forest model was developed based on the DEMs, immune-related DEGs, and metabolites with an AUC value of 0.912. The 14 indicators identified by this model could potentially serve as a set of biomarkers for obesity. The analysis of the inter-omics correlation network found 233 pairs of significant correlations. DEGs BPIFA1, BPI, and SAA1, DEMs Dimethy(tetradecyl)amine, Deoxycholic acid, Pathalic anhydride, and DL-Alanine, and DGMs g_Intestinimonas and g_Turicibacter showed strong connectivity within the network, constituting a large proportion of interactions. CONCLUSION This study presents the first comprehensive description of the multi-omics characteristics of childhood simple obesity, recognizing promising biomarkers. Immune-related markers offer a new perspective for researching the immunological mechanisms underlying obesity and its associated complications. The revealed interactions among these biomarkers contribute to a deeper understanding the intricate biological regulatory networks associated with obesity.
Collapse
Affiliation(s)
- Yi Ren
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Department of Pediatrics, Haikou Hospital of the Maternal and Child Health, Haikou, 570100, China
- Department of Pediatrics, Hainan Modern Women and Children's. Medical, Haikou, 570100, China
| | - Peng Huang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Lu Zhang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Yufen Tang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Siyi He
- Department of Anesthesiology, Hainan General Hospital, Haikou, Hainan, 570311, China
- Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - HaiDan Li
- Department of Pediatrics, Hainan Women and Children's Medical Center, Hainan, 570100, China
| | - XiaoYan Huang
- Department of Pediatrics, Hainan Women and Children's Medical Center, Hainan, 570100, China
| | - Yan Ding
- Department of Dermatology, Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Lingjuan Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Liqun Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Children's Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| | - Xiaojie He
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Laboratory of Pediatric Nephrology, Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
| |
Collapse
|
10
|
Krysa SJ, Brestoff JR. Brown fat fuels the fire in fever. J Lipid Res 2024; 65:100658. [PMID: 39332526 PMCID: PMC11536056 DOI: 10.1016/j.jlr.2024.100658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024] Open
Abstract
Fever is a host-pathogen defense mechanism in which the immune system drives a physiologic increase in core body temperature. For over 50 years, it has been known that the temperature of brown adipose tissue (BAT) is increased during the febrile response. However, recent studies suggested that the primary thermogenic protein Uncoupling protein 1 in brown adipocytes does not contribute to fever induction in mice, casting doubt about the functional contribution of BAT to fever. In a new set of studies, Li et al. (2024) provide compelling evidence that fatty acid oxidation is markedly increased in BAT in a Salmonella infection model of fever and strongly suggest that metabolic adaptation in BAT may play a critical role in the febrile response. This article re-opens the debate about how thermogenic and metabolic programs in BAT contribute to fever and raises new questions about whether BAT contributes to host defense against pathogens.
Collapse
Affiliation(s)
- Samantha J Krysa
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jonathan R Brestoff
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
11
|
Korobkina ED, Calejman CM, Haley JA, Kelly ME, Li H, Gaughan M, Chen Q, Pepper HL, Ahmad H, Boucher A, Fluharty SM, Lin TY, Lotun A, Peura J, Trefely S, Green CR, Vo P, Semenkovich CF, Pitarresi JR, Spinelli JB, Aydemir O, Metallo CM, Lynes MD, Jang C, Snyder NW, Wellen KE, Guertin DA. Brown fat ATP-citrate lyase links carbohydrate availability to thermogenesis and guards against metabolic stress. Nat Metab 2024; 6:2187-2202. [PMID: 39402290 DOI: 10.1038/s42255-024-01143-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 09/16/2024] [Indexed: 11/28/2024]
Abstract
Brown adipose tissue (BAT) engages futile fatty acid synthesis-oxidation cycling, the purpose of which has remained elusive. Here, we show that ATP-citrate lyase (ACLY), which generates acetyl-CoA for fatty acid synthesis, promotes thermogenesis by mitigating metabolic stress. Without ACLY, BAT overloads the tricarboxylic acid cycle, activates the integrated stress response (ISR) and suppresses thermogenesis. ACLY's role in preventing BAT stress becomes critical when mice are weaned onto a carbohydrate-plentiful diet, while removing dietary carbohydrates prevents stress induction in ACLY-deficient BAT. ACLY loss also upregulates fatty acid synthase (Fasn); yet while ISR activation is not caused by impaired fatty acid synthesis per se, deleting Fasn and Acly unlocks an alternative metabolic programme that overcomes tricarboxylic acid cycle overload, prevents ISR activation and rescues thermogenesis. Overall, we uncover a previously unappreciated role for ACLY in mitigating mitochondrial stress that links dietary carbohydrates to uncoupling protein 1-dependent thermogenesis and provides fundamental insight into the fatty acid synthesis-oxidation paradox in BAT.
Collapse
Affiliation(s)
- Ekaterina D Korobkina
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Camila Martinez Calejman
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - John A Haley
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Miranda E Kelly
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Huawei Li
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Maria Gaughan
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Qingbo Chen
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Hannah L Pepper
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Hafsah Ahmad
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Alexander Boucher
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Shelagh M Fluharty
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Te-Yueh Lin
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Anoushka Lotun
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jessica Peura
- Division of Hematology-Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Sophie Trefely
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Courtney R Green
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Paula Vo
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Clay F Semenkovich
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Jason R Pitarresi
- Division of Hematology-Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jessica B Spinelli
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Ozkan Aydemir
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Christian M Metallo
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Molecular and Cellular Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | | | - Cholsoon Jang
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Nathaniel W Snyder
- Aging + Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David A Guertin
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA.
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
12
|
Myers JW, Park WY, Eddie AM, Shinde AB, Prasad P, Murphy AC, Leonard MZ, Pinette JA, Rampy JJ, Montufar C, Shaikh Z, Hickman TT, Reynolds GN, Winn NC, Lantier L, Peck SH, Coate KC, Stein RW, Carrasco N, Calipari ES, McReynolds MR, Zaganjor E. Systemic inhibition of de novo purine biosynthesis prevents weight gain and improves metabolic health by increasing thermogenesis and decreasing food intake. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620705. [PMID: 39553975 PMCID: PMC11566042 DOI: 10.1101/2024.10.28.620705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Objective Obesity is a major health concern, largely because it contributes to type 2 diabetes mellitus (T2DM), cardiovascular disease, and various malignancies. Increase in circulating amino acids and lipids, in part due to adipose dysfunction, have been shown to drive obesity-mediated diseases. Similarly, elevated purines and uric acid, a degradation product of purine metabolism, are found in the bloodstream and in adipose tissue. These metabolic changes are correlated with metabolic syndrome, but little is known about the physiological effects of targeting purine biosynthesis. Methods To determine the effects of purine biosynthesis on organismal health we treated mice with mizoribine, an inhibitor of inosine monophosphate dehydrogenase 1 and 2 (IMPDH1/2), key enzymes in this pathway. Mice were fed either a low-fat (LFD; 13.5% kcal from fat) or a high-fat (HFD; 60% kcal from fat) diet for 30 days during drug or vehicle treatment. We ascertained the effects of mizoribine on weight gain, body composition, food intake and absorption, energy expenditure, and overall metabolic health. Results Mizoribine treatment prevented mice on a HFD from gaining weight, but had no effect on mice on a LFD. Body composition analysis demonstrated that mizoribine significantly reduced fat mass but did not affect lean mass. Although mizoribine had no effect on lipid absorption, food intake was reduced. Furthermore, mizoribine treatment induced adaptive thermogenesis in skeletal muscle by upregulating sarcolipin, a regulator of muscle thermogenesis. While mizoribine-treated mice exhibited less adipose tissue than controls, we did not observe lipotoxicity. Rather, mizoribine-treated mice displayed improved glucose tolerance and reduced ectopic lipid accumulation. Conclusions Inhibiting purine biosynthesis prevents mice on a HFD from gaining weight, and improves their metabolic health, to a significant degree. We also demonstrated that the purine biosynthesis pathway plays a previously unknown role in skeletal muscle thermogenesis. A deeper mechanistic understanding of how purine biosynthesis promotes thermogenesis and decreases food intake may pave the way to new anti-obesity therapies. Crucially, given that many purine inhibitors have been FDA-approved for use in treating various conditions, our results indicate that they may benefit overweight or obese patients.
Collapse
Affiliation(s)
- Jacob W. Myers
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Woo Yong Park
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Alexander M. Eddie
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Abhijit B. Shinde
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Praveena Prasad
- Department of Biochemistry and Molecular Biology, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA
| | - Alexandria C. Murphy
- Department of Biochemistry and Molecular Biology, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA
| | - Michael Z. Leonard
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| | - Julia A. Pinette
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jessica J. Rampy
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT, USA
| | - Claudia Montufar
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Zayedali Shaikh
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Tara T. Hickman
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Garrett N. Reynolds
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Nathan C. Winn
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Louise Lantier
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Mouse Metabolic Phenotyping Center, Nashville, TN, USA
| | - Sun H. Peck
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University School of Engineering, Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Katie C. Coate
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Roland W. Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Nancy Carrasco
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Erin S. Calipari
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| | - Melanie R. McReynolds
- Department of Biochemistry and Molecular Biology, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA
| | - Elma Zaganjor
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Digestive Disease Research Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Diabetes Research and Training Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
13
|
Snyder M, Liu YK, Shang R, Xu H, Thrift C, Chen X, Chen J, Kim KH, Qiu J, Bi P, Tao WA, Kuang S. LETMD1 regulates mitochondrial protein synthesis and import to guard brown fat mitochondrial integrity and function. iScience 2024; 27:110944. [PMID: 39398236 PMCID: PMC11467678 DOI: 10.1016/j.isci.2024.110944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/25/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024] Open
Abstract
Thermogenic brown adipocytes (BAs) catabolize lipids to generate heat, representing powerful agents against the growing global obesity epidemic. We and others reported recently that LETMD1 is a BA-specific protein essential for mitochondrial structure and function, but the mechanisms of action remain unclear. We performed sequential digestion to demonstrate that LETMD1 is a trans-inner mitochondrial membrane protein. We then generated UCP1Cre-driven BA-specific Letmd1 knockout (Letmd1 UKO ) mice to show that Letmd1 UKO leads to protein aggregation, reactive oxidative stress, hyperpolarization, and mitophagy in BAs. We further employed TurboID proximity labeling to identify LETMD1-interacting proteins. Many candidate proteins are associated with mitochondrial ribosomes, protein import machinery, and electron transport chain complexes (ETC-I and ETC-IV). Using quantitative proteomics, we confirmed the elevated aggregations of ETC and mitochondrial ribosomal proteins, impairing mitochondrial protein synthesis in the Letmd1 UKO BAs. Therefore, LETMD1 may function to maintain mitochondrial proteostasis through regulating import of nuclear-encoded proteins and local protein translation in brown fat mitochondria.
Collapse
Affiliation(s)
- Madigan Snyder
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Yi-Kai Liu
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Renjie Shang
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Haowei Xu
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Charlie Thrift
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Xiyue Chen
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Jingjuan Chen
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
- Department of Orthopaedic Surgery, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Kun Ho Kim
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Jiamin Qiu
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Pengpeng Bi
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - W. Andy Tao
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907, USA
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
- Department of Orthopaedic Surgery, School of Medicine, Duke University, Durham, NC 27710, USA
| |
Collapse
|
14
|
Chen R, Cheng T, Xie S, Sun X, Chen M, Zhao S, Ruan Q, Ni X, Rao M, Quan X, Chen K, Zhang S, Cheng T, Xu Y, Chen Y, Yang Y, Cao Y. Effective Prevention and Treatment of Acute Leukemias in Mice by Activation of Thermogenic Adipose Tissues. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402332. [PMID: 39049685 PMCID: PMC11481385 DOI: 10.1002/advs.202402332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 07/09/2024] [Indexed: 07/27/2024]
Abstract
Acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) are common hematological malignancies in adults. Despite considerable research advances, the development of standard therapies, supportive care, and prognosis for the majority of AML and ALL patients remains poor and the development of new effective therapy is urgently needed. Here, it is reported that activation of thermogenic adipose tissues (TATs) by cold exposure or β3-adrenergic receptor agonists markedly alleviated the development and progression of AML and ALL in mouse leukemia models. TAT activation (TATA) monotherapy substantially reduces leukemic cells in bone marrow and peripheral blood, and suppresses leukemic cell invasion, including hepatomegaly and splenomegaly. Notably, TATA therapy prolongs the survivals of AML- and ALL-bearing mice. Surgical removal of thermogenic brown adipose tissue (BAT) or genetic deletion of uncoupling protein 1 (UCP1) largely abolishes the TATA-mediated anti-leukemia effects. Metabolomic pathway analysis demonstrates that glycolytic metabolism, which is essential for anabolic leukemic cell growth, is severely impaired in TATA-treated leukemic cells. Moreover, a combination of TATA therapy with chemotherapy produces enhanced anti-leukemic effects and reduces chemotoxicity. These data provide a new TATA-based therapeutic paradigm for the effective treatment of AML, ALL, and likely other types of hematological malignancies.
Collapse
Affiliation(s)
- Ruibo Chen
- Department of Cellular and Genetic MedicineSchool of Basic Medical SciencesFudan UniversityShanghai200032China
| | - Tianran Cheng
- State Key Laboratory of Experimental HematologyNational Clinical Research Center for Blood DiseasesHaihe Laboratory of Cell EcosystemInstitute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300020China
| | - Sisi Xie
- Department of CardiologyBasic Scientific Research CenterLongyan First Hospital Affiliated to Fujian Medical UniversityLongyan364000China
| | - Xiaoting Sun
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)School of Pharmaceutical ScienceWenzhou Medical UniversityWenzhou325035China
| | - Mingjia Chen
- Department of Cellular and Genetic MedicineSchool of Basic Medical SciencesFudan UniversityShanghai200032China
| | - Shumin Zhao
- Department of CardiologyBasic Scientific Research CenterLongyan First Hospital Affiliated to Fujian Medical UniversityLongyan364000China
| | - Qingyan Ruan
- Department of CardiologyBasic Scientific Research CenterLongyan First Hospital Affiliated to Fujian Medical UniversityLongyan364000China
| | - Xiaolei Ni
- Department of CardiologyBasic Scientific Research CenterLongyan First Hospital Affiliated to Fujian Medical UniversityLongyan364000China
| | - Mei Rao
- Department of CardiologyBasic Scientific Research CenterLongyan First Hospital Affiliated to Fujian Medical UniversityLongyan364000China
| | - Xinyi Quan
- Department of Cellular and Genetic MedicineSchool of Basic Medical SciencesFudan UniversityShanghai200032China
| | - Kaiwen Chen
- Department of Cellular and Genetic MedicineSchool of Basic Medical SciencesFudan UniversityShanghai200032China
| | - Shiyue Zhang
- State Key Laboratory of Experimental HematologyNational Clinical Research Center for Blood DiseasesHaihe Laboratory of Cell EcosystemInstitute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300020China
| | - Tao Cheng
- State Key Laboratory of Experimental HematologyNational Clinical Research Center for Blood DiseasesHaihe Laboratory of Cell EcosystemInstitute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300020China
| | - Yuanfu Xu
- State Key Laboratory of Experimental HematologyNational Clinical Research Center for Blood DiseasesHaihe Laboratory of Cell EcosystemInstitute of Hematology & Blood Diseases HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjin300020China
| | - Yuguo Chen
- Department of Emergency MedicineShandong Provincial Clinical Research Center for Emergency and Critical Care MedicineMedical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care MedicineChina’s Ministry of EducationNMPA Key Laboratory for Clinical Research and Evaluation of Innovative DrugShandong International Cooperative Laboratory for Emergency and Critical Care MedicineQilu Hospital of Shandong UniversityJinan250012China
| | - Yunlong Yang
- Department of Cellular and Genetic MedicineSchool of Basic Medical SciencesFudan UniversityShanghai200032China
| | - Yihai Cao
- Department of MicrobiologyTumor and Cell BiologyKarolinska InstitutetSolna17165Sweden
| |
Collapse
|
15
|
Vargas-Castillo A, Sun Y, Smythers AL, Grauvogel L, Dumesic PA, Emont MP, Tsai LT, Rosen ED, Zammit NW, Shaffer SM, Ordonez M, Chouchani ET, Gygi SP, Wang T, Sharma AK, Balaz M, Wolfrum C, Spiegelman BM. Development of a functional beige fat cell line uncovers independent subclasses of cells expressing UCP1 and the futile creatine cycle. Cell Metab 2024; 36:2146-2155.e5. [PMID: 39084217 DOI: 10.1016/j.cmet.2024.07.002] [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: 12/19/2023] [Revised: 04/30/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024]
Abstract
Although uncoupling protein 1 (UCP1) is established as a major contributor to adipose thermogenesis, recent data have illustrated an important role for alternative pathways, particularly the futile creatine cycle (FCC). How these pathways co-exist in cells and tissues has not been explored. Beige cell adipogenesis occurs in vivo but has been difficult to model in vitro; here, we describe the development of a murine beige cell line that executes a robust respiratory response, including uncoupled respiration and the FCC. The key FCC enzyme, tissue-nonspecific alkaline phosphatase (TNAP), is localized almost exclusively to mitochondria in these cells. Surprisingly, single-cell cloning from this cell line shows that cells with the highest levels of UCP1 express little TNAP, and cells with the highest expression of TNAP express little UCP1. Immunofluorescence analysis of subcutaneous fat from cold-exposed mice confirms that the highest levels of these critical thermogenic components are expressed in distinct fat cell populations.
Collapse
Affiliation(s)
- Ariana Vargas-Castillo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Yizhi Sun
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Amanda L Smythers
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Louisa Grauvogel
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Phillip A Dumesic
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Margo P Emont
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Linus T Tsai
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Evan D Rosen
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nathan W Zammit
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Sydney M Shaffer
- Department of Pathology and Laboratory Medicine and the Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Martha Ordonez
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Edward T Chouchani
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Tongtong Wang
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Anand K Sharma
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Miroslav Balaz
- Laboratory of Cellular and Molecular Metabolism, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Christian Wolfrum
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Wang T, Sharma AK, Wu C, Maushart CI, Ghosh A, Yang W, Stefanicka P, Kovanicova Z, Ukropec J, Zhang J, Arnold M, Klug M, De Bock K, Schneider U, Popescu C, Zheng B, Ding L, Long F, Dewal RS, Moser C, Sun W, Dong H, Takes M, Suelberg D, Mameghani A, Nocito A, Zech CJ, Chirindel A, Wild D, Burger IA, Schön MR, Dietrich A, Gao M, Heine M, Sun Y, Vargas-Castillo A, Søberg S, Scheele C, Balaz M, Blüher M, Betz MJ, Spiegelman BM, Wolfrum C. Single-nucleus transcriptomics identifies separate classes of UCP1 and futile cycle adipocytes. Cell Metab 2024; 36:2130-2145.e7. [PMID: 39084216 DOI: 10.1016/j.cmet.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/22/2024] [Accepted: 07/08/2024] [Indexed: 08/02/2024]
Abstract
Adipose tissue can recruit catabolic adipocytes that utilize chemical energy to dissipate heat. This process occurs either by uncoupled respiration through uncoupling protein 1 (UCP1) or by utilizing ATP-dependent futile cycles (FCs). However, it remains unclear how these pathways coexist since both processes rely on the mitochondrial membrane potential. Utilizing single-nucleus RNA sequencing to deconvolute the heterogeneity of subcutaneous adipose tissue in mice and humans, we identify at least 2 distinct subpopulations of beige adipocytes: FC-adipocytes and UCP1-beige adipocytes. Importantly, we demonstrate that the FC-adipocyte subpopulation is highly metabolically active and utilizes FCs to dissipate energy, thus contributing to thermogenesis independent of Ucp1. Furthermore, FC-adipocytes are important drivers of systemic energy homeostasis and linked to glucose metabolism and obesity resistance in humans. Taken together, our findings identify a noncanonical thermogenic adipocyte subpopulation, which could be an important regulator of energy homeostasis in mammals.
Collapse
Affiliation(s)
- Tongtong Wang
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Anand Kumar Sharma
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Chunyan Wu
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Claudia Irene Maushart
- Department of Endocrinology, Diabetes, and Metabolism, University Hospital of Basel and University of Basel, Basel, Switzerland
| | - Adhideb Ghosh
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Wu Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Patrik Stefanicka
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine and University Hospital, Comenius University in Bratislava, Bratislava, Slovakia
| | - Zuzana Kovanicova
- Institute of Experimental Endocrinology, Biomedical Research Center at the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Jozef Ukropec
- Institute of Experimental Endocrinology, Biomedical Research Center at the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Jing Zhang
- Laboratory of Exercise and Health, Health Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, Switzerland
| | - Myrtha Arnold
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Manuel Klug
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Katrien De Bock
- Laboratory of Exercise and Health, Health Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, Switzerland
| | - Ulrich Schneider
- Department of Surgery, Cantonal Hospital of Baden, Im Ergel 1, 5404 Baden, Switzerland
| | - Cristina Popescu
- Department of Nuclear Medicine, Cantonal Hospital of Baden, Im Ergel 1, 5404 Baden, Switzerland
| | - Bo Zheng
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Lianggong Ding
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Fen Long
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Revati Sumukh Dewal
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Caroline Moser
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Wenfei Sun
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Hua Dong
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Martin Takes
- Department of Radiology and Nuclear Medicine, University Hospital of Basel, Basel, Switzerland
| | - Dominique Suelberg
- Department of Surgery, Cantonal Hospital of Baden, Im Ergel 1, 5404 Baden, Switzerland
| | - Alexander Mameghani
- Department of Surgery, Cantonal Hospital of Baden, Im Ergel 1, 5404 Baden, Switzerland
| | - Antonio Nocito
- Department of Surgery, Cantonal Hospital of Baden, Im Ergel 1, 5404 Baden, Switzerland
| | - Christoph Johannes Zech
- Department of Radiology and Nuclear Medicine, University Hospital of Basel, Basel, Switzerland
| | - Alin Chirindel
- Department of Radiology and Nuclear Medicine, University Hospital of Basel, Basel, Switzerland
| | - Damian Wild
- Department of Radiology and Nuclear Medicine, University Hospital of Basel, Basel, Switzerland
| | - Irene A Burger
- Department of Nuclear Medicine, Cantonal Hospital of Baden, Im Ergel 1, 5404 Baden, Switzerland; Department of Nuclear Medicine, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Michael R Schön
- Städtisches Klinikum Karlsruhe, Clinic of Visceral Surgery, Karlsruhe, Germany
| | - Arne Dietrich
- Clinic for Visceral, Transplant and Thoracic and Vascular Surgery, University Hospital Leipzig, Liebigstrasse 20, 04103 Leipzig, Germany
| | - Min Gao
- Department of Pharmacy, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Yizhi Sun
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Ariana Vargas-Castillo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Susanna Søberg
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; The Center of Inflammation and Metabolism and the Center for Physical Activity Research, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Camilla Scheele
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; The Center of Inflammation and Metabolism and the Center for Physical Activity Research, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Miroslav Balaz
- Institute of Experimental Endocrinology, Biomedical Research Center at the Slovak Academy of Sciences, Bratislava, Slovakia; Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia
| | - Matthias Blüher
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Germany & Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital, Leipzig, Germany.
| | - Matthias Johannes Betz
- Department of Endocrinology, Diabetes, and Metabolism, University Hospital of Basel and University of Basel, Basel, Switzerland.
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Christian Wolfrum
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|
17
|
Rodriguez-Muñoz A, Motahari-Rad H, Martin-Chaves L, Benitez-Porres J, Rodriguez-Capitan J, Gonzalez-Jimenez A, Insenser M, Tinahones FJ, Murri M. A Systematic Review of Proteomics in Obesity: Unpacking the Molecular Puzzle. Curr Obes Rep 2024; 13:403-438. [PMID: 38703299 PMCID: PMC11306592 DOI: 10.1007/s13679-024-00561-4] [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] [Accepted: 03/14/2024] [Indexed: 05/06/2024]
Abstract
PURPOSE OF REVIEW The present study aims to review the existing literature to identify pathophysiological proteins in obesity by conducting a systematic review of proteomics studies. Proteomics may reveal the mechanisms of obesity development and clarify the links between obesity and related diseases, improving our comprehension of obesity and its clinical implications. RECENT FINDINGS Most of the molecular events implicated in obesity development remain incomplete. Proteomics stands as a powerful tool for elucidating the intricate interactions among proteins in the context of obesity. This methodology has the potential to identify proteins involved in pathological processes and to evaluate changes in protein abundance during obesity development, contributing to the identification of early disease predisposition, monitoring the effectiveness of interventions and improving disease management overall. Despite many non-targeted proteomic studies exploring obesity, a comprehensive and up-to-date systematic review of the molecular events implicated in obesity development is lacking. The lack of such a review presents a significant challenge for researchers trying to interpret the existing literature. This systematic review was conducted following the PRISMA guidelines and included sixteen human proteomic studies, each of which delineated proteins exhibiting significant alterations in obesity. A total of 41 proteins were reported to be altered in obesity by at least two or more studies. These proteins were involved in metabolic pathways, oxidative stress responses, inflammatory processes, protein folding, coagulation, as well as structure/cytoskeleton. Many of the identified proteomic biomarkers of obesity have also been reported to be dysregulated in obesity-related disease. Among them, seven proteins, which belong to metabolic pathways (aldehyde dehydrogenase and apolipoprotein A1), the chaperone family (albumin, heat shock protein beta 1, protein disulfide-isomerase A3) and oxidative stress and inflammation proteins (catalase and complement C3), could potentially serve as biomarkers for the progression of obesity and the development of comorbidities, contributing to personalized medicine in the field of obesity. Our systematic review in proteomics represents a substantial step forward in unravelling the complexities of protein alterations associated with obesity. It provides valuable insights into the pathophysiological mechanisms underlying obesity, thereby opening avenues for the discovery of potential biomarkers and the development of personalized medicine in obesity.
Collapse
Affiliation(s)
- Alba Rodriguez-Muñoz
- Endocrinology and Nutrition UGC, Hospital Universitario Virgen de La Victoria, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Clínico Virgen de La Victoria, Málaga, Spain
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Málaga, Spain
| | - Hanieh Motahari-Rad
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Laura Martin-Chaves
- Heart Area, Hospital Universitario Virgen de La Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
- Department of Dermatology and Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Javier Benitez-Porres
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Clínico Virgen de La Victoria, Málaga, Spain
- Department of Human Physiology, Physical Education and Sport, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Jorge Rodriguez-Capitan
- Heart Area, Hospital Universitario Virgen de La Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
- Biomedical Research Network Center for Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | | | - Maria Insenser
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology & Nutrition, Hospital Universitario Ramón y Cajal & Universidad de Alcalá & Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain.
| | - Francisco J Tinahones
- Endocrinology and Nutrition UGC, Hospital Universitario Virgen de La Victoria, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Clínico Virgen de La Victoria, Málaga, Spain
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Málaga, Spain
- Department of Dermatology and Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Mora Murri
- Endocrinology and Nutrition UGC, Hospital Universitario Virgen de La Victoria, Málaga, Spain.
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Clínico Virgen de La Victoria, Málaga, Spain.
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Málaga, Spain.
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
18
|
Li SJ, Wei JQ, Kang YY, Wang RQ, Rong WW, Zhao JJ, Deng QW, Gao PJ, Li XD, Wang JG. Natriuretic peptide receptor-C perturbs mitochondrial respiration in white adipose tissue. J Lipid Res 2024; 65:100623. [PMID: 39154732 PMCID: PMC11418126 DOI: 10.1016/j.jlr.2024.100623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/15/2024] [Accepted: 08/11/2024] [Indexed: 08/20/2024] Open
Abstract
Natriuretic peptide receptor-C (NPR-C) is highly expressed in adipose tissues and regulates obesity-related diseases; however, the detailed mechanism remains unknown. In this research, we aimed to explore the potential role of NPR-C in cold exposure and high-fat/high-sugar (HF/HS) diet-induced metabolic changes, especially in regulating white adipose tissue (WAT) mitochondrial function. Our findings showed that NPR-C expression, especially in epididymal WAT (eWAT), was reduced after cold exposure. Global Npr3 (gene encoding NPR-C protein) deficiency led to reduced body weight, increased WAT browning, thermogenesis, and enhanced expression of genes related to mitochondrial biogenesis. RNA-sequencing of eWAT showed that Npr3 deficiency enhanced the expression of mitochondrial respiratory chain complex genes and promoted mitochondrial oxidative phosphorylation in response to cold exposure. In addition, Npr3 KO mice were able to resist obesity induced by HF/HS diet. Npr3 knockdown in stromal vascular fraction (SVF)-induced white adipocytes promoted the expression of proliferator-activated receptor gamma coactivator 1α (PGC1α), uncoupling protein one (UCP1), and mitochondrial respiratory chain complexes. Mechanistically, NPR-C inhibited cGMP and calcium signaling in an NPR-B-dependent manner but suppressed cAMP signaling in an NPR-B-independent manner. Moreover, Npr3 knockdown induced browning via AKT and p38 pathway activation, which were attenuated by Npr2 knockdown. Importantly, treatment with the NPR-C-specific antagonist, AP-811, decreased WAT mass and increased PGC-1α, UCP1, and mitochondrial complex expression. Our findings reveal that NPR-C deficiency enhances mitochondrial function and energy expenditure in white adipose tissue, contributing to improved metabolic health and resistance to obesity.
Collapse
Affiliation(s)
- Shi-Jin Li
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Jin-Qiu Wei
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan-Yuan Kang
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui-Qi Wang
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Wu-Wei Rong
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia-Jia Zhao
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian-Wan Deng
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping-Jin Gao
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Dong Li
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ji-Guang Wang
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Cai Z, Zhong Q, Zhang D, Feng Y, Wang Q, Yang Y, Xu Y, Liang C, Liu Z, Cai K. Z-Spectral MRI Quantifies the Mass and Metabolic Activity of Adipose Tissues With Fat-Water-Fraction and Amide-Proton-Transfer Contrasts. J Magn Reson Imaging 2024. [PMID: 39215496 DOI: 10.1002/jmri.29598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Brown adipose tissue (BAT) is metabolically activatable and plays an important role in obesity and metabolic diseases. With reduced fat-water-fraction (FWF) compared with white adipose tissue (WAT), BAT mass and its functional activation may be quantified with Z-spectra MRI, with built-in FWF and the metabolic amide proton transfer (APT) contrasts. PURPOSE To investigate if Z-spectral MRI can quantify the mass and metabolic activity of adipose tissues. STUDY TYPE Prospective. SUBJECTS Seven groups of 8-week-old male rats, including two groups (n = 7 per group) for in vivo MRI study and five groups (n = 5 per group) for ex vivo validation; 12 young and healthy volunteers with 6 male and 6 female. FIELD STRENGTH/SEQUENCE The 7 T small animal and 3 T clinical systems, T2-weighted imaging, Rapid Acquisition with Relaxation Enhancement (RARE) readout based chemical exchange saturation transfer (CEST) Z-spectral MRI sequence. ASSESSMENT Quantified FWF and APT from Z-spectra in rats before and after norepinephrine (NE) stimulation and in healthy human subjects; ex vivo measurements of total proteins in BAT from rats. STATISTICAL TESTS Two-tailed unpaired Student's t-tests and repeated measures ANOVA. P-value <0.05 was considered significant. RESULTS Decreased FWF (from 39.6% ± 7.2% before NE injection to 16.4% ± 7.2% 120 minutes after NE injection, P < 0.0001) and elevated APT (from 1.1% ± 0.5% before NE injection to 2.9% ± 0.5% 120 minutes after NE injection, P < 0.0001) signals in BAT were observed with in vivo Z-spectral MRI in rats injected with NE at 7 T MRI. At clinical 3 T, Z-spectral MRI was used to quantify the FWF (58.5% ± 7.2% in BAT and 73.7% ± 6.5% in WAT with P < 0.0001) and APT (2.6% ± 0.8% in BAT and 0.9% ± 0.3% in WAT with P < 0.0001) signals in healthy volunteers. APT signals of BAT were negatively correlated with the BMI in humans (r = 0.71). DATA CONCLUSION Endogenous Z-spectral MRI was demonstrated to simultaneously quantify BAT mass and function based on its FWF and APT contrasts. LEVEL OF EVIDENCE: 2 TECHNICAL EFFICACY STAGE 1.
Collapse
Affiliation(s)
- Zimeng Cai
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Qiaoling Zhong
- Department of Radiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Daming Zhang
- State Key Laboratory of Bioactive Molecules and Draggability Assessment, The Biomedical Translational Research Institute, Health Science Center, School of Medicine, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, China
| | - Yanqiu Feng
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Image Processing & Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence & Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, China
- Department of Radiology, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, Foshan, China
| | - Qian Wang
- State Key Laboratory of Bioactive Molecules and Draggability Assessment, The Biomedical Translational Research Institute, Health Science Center, School of Medicine, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, China
| | - Yuanbo Yang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | | | - Changhong Liang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Zaiyi Liu
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Kejia Cai
- Radiology Department, University of Illinois at Chicago, Chicago, Illinois, USA
- Biomedical Engineering Department, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
20
|
Takeda Y, Dai P. Functional roles of pantothenic acid, riboflavin, thiamine, and choline in adipocyte browning in chemically induced human brown adipocytes. Sci Rep 2024; 14:18252. [PMID: 39107469 PMCID: PMC11303702 DOI: 10.1038/s41598-024-69364-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024] Open
Abstract
Brown fat is a therapeutic target for the treatment of obesity-associated metabolic diseases. However, nutritional intervention strategies for increasing the mass and activity of human brown adipocytes have not yet been established. To identify vitamins required for brown adipogenesis and adipocyte browning, chemical compound-induced brown adipocytes (ciBAs) were converted from human dermal fibroblasts under serum-free and vitamin-free conditions. Choline was found to be essential for adipogenesis. Additional treatment with pantothenic acid (PA) provided choline-induced immature adipocytes with browning properties and metabolic maturation, including uncoupling protein 1 (UCP1) expression, lipolysis, and mitochondrial respiration. However, treatment with high PA concentrations attenuated these effects along with decreased glycolysis. Transcriptome analysis showed that a low PA concentration activated metabolic genes, including the futile creatine cycle-related thermogenic genes, which was reversed by a high PA concentration. Riboflavin treatment suppressed thermogenic gene expression and increased lipolysis, implying a metabolic pathway different from that of PA. Thiamine treatment slightly activated thermogenic genes along with decreased glycolysis. In summary, our results suggest that specific B vitamins and choline are uniquely involved in the regulation of adipocyte browning via cellular energy metabolism in a concentration-dependent manner.
Collapse
Affiliation(s)
- Yukimasa Takeda
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan.
| | - Ping Dai
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-Cho, Kawaramachi-Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan.
| |
Collapse
|
21
|
He L, Lin J, Lu S, Li H, Chen J, Wu X, Yan Q, Liu H, Li H, Shi Y. CKB Promotes Mitochondrial ATP Production by Suppressing Permeability Transition Pore. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403093. [PMID: 38896801 PMCID: PMC11336976 DOI: 10.1002/advs.202403093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/04/2024] [Indexed: 06/21/2024]
Abstract
Creatine kinases are essential for maintaining cellular energy balance by facilitating the reversible transfer of a phosphoryl group from ATP to creatine, however, their role in mitochondrial ATP production remains unknown. This study shows creatine kinases, including CKMT1A, CKMT1B, and CKB, are highly expressed in cells relying on the mitochondrial F1F0 ATP synthase for survival. Interestingly, silencing CKB, but not CKMT1A or CKMT1B, leads to a loss of sensitivity to the inhibition of F1F0 ATP synthase in these cells. Mechanistically, CKB promotes mitochondrial ATP but reduces glycolytic ATP production by suppressing mitochondrial calcium (mCa2+) levels, thereby preventing the activation of mitochondrial permeability transition pore (mPTP) and ensuring efficient mitochondrial ATP generation. Further, CKB achieves this regulation by suppressing mCa2+ levels through the inhibition of AKT activity. Notably, the CKB-AKT signaling axis boosts mitochondrial ATP production in cancer cells growing in a mouse tumor model. Moreover, this study also uncovers a decline in CKB expression in peripheral blood mononuclear cells with aging, accompanied by an increase in AKT signaling in these cells. These findings thus shed light on a novel signaling pathway involving CKB that directly regulates mitochondrial ATP production, potentially playing a role in both pathological and physiological conditions.
Collapse
Affiliation(s)
- Le He
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Jianghua Lin
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Shaojuan Lu
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Hao Li
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Jie Chen
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Xinyi Wu
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Qixin Yan
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Hailiang Liu
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
- State Key Laboratory of Cardiology and Medical Innovation CenterShanghai East HospitalSchool of MedicineTongji UniversityShanghai200123China
| | - Hui Li
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Yufeng Shi
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationTongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| |
Collapse
|
22
|
Varghese A, Gusarov I, Gamallo-Lana B, Dolgonos D, Mankan Y, Shamovsky I, Phan M, Jones R, Gomez-Jenkins M, White E, Wang R, Jones D, Papagiannakopoulos T, Pacold ME, Mar AC, Littman DR, Nudler E. Unraveling cysteine deficiency-associated rapid weight loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.30.605703. [PMID: 39131293 PMCID: PMC11312522 DOI: 10.1101/2024.07.30.605703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Forty percent of the US population and 1 in 6 individuals worldwide are obese, and the incidence of this disease is surging globally1,2. Various dietary interventions, including carbohydrate and fat restriction, and more recently amino acid restriction, have been explored to combat this epidemic3-6. We sought to investigate the impact of removing individual amino acids on the weight profiles of mice. Compared to essential amino acid restriction, induction of conditional cysteine restriction resulted in the most dramatic weight loss, amounting to 20% within 3 days and 30% within one week, which was readily reversed. This weight loss occurred despite the presence of substantial cysteine reserves stored in glutathione (GSH) across various tissues7. Further analysis demonstrated that the weight reduction primarily stemmed from an increase in the utilization of fat mass, while locomotion, circadian rhythm and histological appearance of multiple other tissues remained largely unaffected. Cysteine deficiency activated the integrated stress response (ISR) and NRF2-mediated oxidative stress response (OSR), which amplify each other, leading to the induction of GDF15 and FGF21, hormones associated with increased lipolysis, energy homeostasis and food aversion8-10. We additionally observed rapid tissue coenzyme A (CoA) depletion, resulting in energetically inefficient anaerobic glycolysis and TCA cycle, with sustained urinary excretion of pyruvate, orotate, citrate, α-ketoglutarate, nitrogen rich compounds and amino acids. In summary, our investigation highlights that cysteine restriction, by depleting GSH and CoA, exerts a maximal impact on weight loss, metabolism, and stress signaling compared to other amino acid restrictions. These findings may pave the way for innovative strategies for addressing a range of metabolic diseases and the growing obesity crisis.
Collapse
Affiliation(s)
- Alan Varghese
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Ivan Gusarov
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Begoña Gamallo-Lana
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU School of Medicine, New York, NY 10016, USA
| | - Daria Dolgonos
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Yatin Mankan
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Ilya Shamovsky
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Mydia Phan
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Rebecca Jones
- Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Maria Gomez-Jenkins
- Rutgers Cancer Institute, Rutgers University, New Brunswick, NJ 08901, USA and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | - Eileen White
- Rutgers Cancer Institute, Rutgers University, New Brunswick, NJ 08901, USA and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
| | - Rui Wang
- Department of Biology, York University, Toronto, Ontario, M3J 1P3, Canada
| | - Drew Jones
- Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Thales Papagiannakopoulos
- Department of Pathology, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Michael E Pacold
- Department of Radiation Oncology and Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York, NY 10016, USA
| | - Adam C Mar
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU School of Medicine, New York, NY 10016, USA
| | - Dan R Littman
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Howard Hughes Medical Institute, New York, NY 10016, USA
| | - Evgeny Nudler
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Howard Hughes Medical Institute, New York, NY 10016, USA
| |
Collapse
|
23
|
Prapaharan B, Lea M, Beaudry JL. Weighing in on the role of brown adipose tissue for treatment of obesity. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:13157. [PMID: 39087083 PMCID: PMC11290130 DOI: 10.3389/jpps.2024.13157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024]
Abstract
Brown adipose tissue (BAT) activation is an emerging target for obesity treatments due to its thermogenic properties stemming from its ability to shuttle energy through uncoupling protein 1 (Ucp1). Recent rodent studies show how BAT and white adipose tissue (WAT) activity can be modulated to increase the expression of thermogenic proteins. Consequently, these alterations enable organisms to endure cold-temperatures and elevate energy expenditure, thereby promoting weight loss. In humans, BAT is less abundant in obese subjects and impacts of thermogenesis are less pronounced, bringing into question whether energy expending properties of BAT seen in rodents can be translated to human models. Our review will discuss pharmacological, hormonal, bioactive, sex-specific and environmental activators and inhibitors of BAT to determine the potential for BAT to act as a therapeutic strategy. We aim to address the feasibility of utilizing BAT modulators for weight reduction in obese individuals, as recent studies suggest that BAT's contributions to energy expenditure along with Ucp1-dependent and -independent pathways may or may not rectify energy imbalance characteristic of obesity.
Collapse
Affiliation(s)
| | | | - Jacqueline L. Beaudry
- Temerty Faculty of Medicine, Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
24
|
Jamerson LE, Bradshaw PC. The Roles of White Adipose Tissue and Liver NADPH in Dietary Restriction-Induced Longevity. Antioxidants (Basel) 2024; 13:820. [PMID: 39061889 PMCID: PMC11273496 DOI: 10.3390/antiox13070820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Dietary restriction (DR) protocols frequently employ intermittent fasting. Following a period of fasting, meal consumption increases lipogenic gene expression, including that of NADPH-generating enzymes that fuel lipogenesis in white adipose tissue (WAT) through the induction of transcriptional regulators SREBP-1c and CHREBP. SREBP-1c knockout mice, unlike controls, did not show an extended lifespan on the DR diet. WAT cytoplasmic NADPH is generated by both malic enzyme 1 (ME1) and the pentose phosphate pathway (PPP), while liver cytoplasmic NADPH is primarily synthesized by folate cycle enzymes provided one-carbon units through serine catabolism. During the daily fasting period of the DR diet, fatty acids are released from WAT and are transported to peripheral tissues, where they are used for beta-oxidation and for phospholipid and lipid droplet synthesis, where monounsaturated fatty acids (MUFAs) may activate Nrf1 and inhibit ferroptosis to promote longevity. Decreased WAT NADPH from PPP gene knockout stimulated the browning of WAT and protected from a high-fat diet, while high levels of NADPH-generating enzymes in WAT and macrophages are linked to obesity. But oscillations in WAT [NADPH]/[NADP+] from feeding and fasting cycles may play an important role in maintaining metabolic plasticity to drive longevity. Studies measuring the WAT malate/pyruvate as a proxy for the cytoplasmic [NADPH]/[NADP+], as well as studies using fluorescent biosensors expressed in the WAT of animal models to monitor the changes in cytoplasmic [NADPH]/[NADP+], are needed during ad libitum and DR diets to determine the changes that are associated with longevity.
Collapse
Affiliation(s)
| | - Patrick C. Bradshaw
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| |
Collapse
|
25
|
Cai Z, Zhong Q, Feng Y, Wang Q, Zhang Z, Wei C, Yin Z, Liang C, Liew CW, Kazak L, Cypess AM, Liu Z, Cai K. Non-invasive mapping of brown adipose tissue activity with magnetic resonance imaging. Nat Metab 2024; 6:1367-1379. [PMID: 39054361 PMCID: PMC11272596 DOI: 10.1038/s42255-024-01082-z] [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: 08/22/2023] [Accepted: 06/14/2024] [Indexed: 07/27/2024]
Abstract
Thermogenic brown adipose tissue (BAT) has a positive impact on whole-body metabolism. However, in vivo mapping of BAT activity typically relies on techniques involving ionizing radiation, such as [18F]fluorodeoxyglucose ([18F]FDG) positron emission tomography (PET) and computed tomography (CT). Here we report a noninvasive metabolic magnetic resonance imaging (MRI) approach based on creatine chemical exchange saturation transfer (Cr-CEST) contrast to assess in vivo BAT activity in rodents and humans. In male rats, a single dose of the β3-adrenoceptor agonist (CL 316,243) or norepinephrine, as well as cold exposure, triggered a robust elevation of the Cr-CEST MRI signal, which was consistent with the [18F]FDG PET and CT data and 1H nuclear magnetic resonance measurements of creatine concentration in BAT. We further show that Cr-CEST MRI detects cold-stimulated BAT activation in humans (both males and females) using a 3T clinical scanner, with data-matching results from [18F]FDG PET and CT measurements. This study establishes Cr-CEST MRI as a promising noninvasive and radiation-free approach for in vivo mapping of BAT activity.
Collapse
Affiliation(s)
- Zimeng Cai
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Qiaoling Zhong
- Department of Radiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yanqiu Feng
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Image Processing & Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence & Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, China
| | - Qian Wang
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
| | - Zuoman Zhang
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Cailv Wei
- School of Medicine, Shenzhen Campus, Sun Yat-sen University, Shenzhen, China
| | - Zhinan Yin
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, China
| | - Changhong Liang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Chong Wee Liew
- Physiology and Biophysics Department, University of Illinois at Chicago, Chicago, IL, USA
| | - Lawrence Kazak
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, Intramural Research Program, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China.
| | - Kejia Cai
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
26
|
Yang S, Liu Y, Wu X, Zhu R, Sun Y, Zou S, Zhang D, Yang X. Molecular Regulation of Thermogenic Mechanisms in Beige Adipocytes. Int J Mol Sci 2024; 25:6303. [PMID: 38928011 PMCID: PMC11203837 DOI: 10.3390/ijms25126303] [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: 05/10/2024] [Revised: 06/02/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Adipose tissue is conventionally recognized as a metabolic organ responsible for storing energy. However, a proportion of adipose tissue also functions as a thermogenic organ, contributing to the inhibition of weight gain and prevention of metabolic diseases. In recent years, there has been significant progress in the study of thermogenic fats, particularly brown adipose tissue (BAT). Despite this progress, the mechanism underlying thermogenesis in beige adipose tissue remains highly controversial. It is widely acknowledged that beige adipose tissue has three additional thermogenic mechanisms in addition to the conventional UCP1-dependent thermogenesis: Ca2+ cycling thermogenesis, creatine substrate cycling thermogenesis, and triacylglycerol/fatty acid cycling thermogenesis. This paper delves into these three mechanisms and reviews the latest advancements in the molecular regulation of thermogenesis from the molecular genetic perspective. The objective of this review is to provide readers with a foundation of knowledge regarding the beige fats and a foundation for future research into the mechanisms of this process, which may lead to the development of new strategies for maintaining human health.
Collapse
Affiliation(s)
- Siqi Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China; (S.Y.); (Y.L.); (X.W.); (R.Z.); (Y.S.); (S.Z.)
| | - Yingke Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China; (S.Y.); (Y.L.); (X.W.); (R.Z.); (Y.S.); (S.Z.)
| | - Xiaoxu Wu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China; (S.Y.); (Y.L.); (X.W.); (R.Z.); (Y.S.); (S.Z.)
| | - Rongru Zhu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China; (S.Y.); (Y.L.); (X.W.); (R.Z.); (Y.S.); (S.Z.)
| | - Yuanlu Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China; (S.Y.); (Y.L.); (X.W.); (R.Z.); (Y.S.); (S.Z.)
| | - Shuoya Zou
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China; (S.Y.); (Y.L.); (X.W.); (R.Z.); (Y.S.); (S.Z.)
| | - Dongjie Zhang
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Xiuqin Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China; (S.Y.); (Y.L.); (X.W.); (R.Z.); (Y.S.); (S.Z.)
| |
Collapse
|
27
|
Keipert S, Gaudry MJ, Kutschke M, Keuper M, Dela Rosa MAS, Cheng Y, Monroy Kuhn JM, Laterveer R, Cotrim CA, Giere P, Perocchi F, Feederle R, Crichton PG, Lutter D, Jastroch M. Two-stage evolution of mammalian adipose tissue thermogenesis. Science 2024; 384:1111-1117. [PMID: 38843333 DOI: 10.1126/science.adg1947] [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: 07/02/2023] [Accepted: 04/08/2024] [Indexed: 06/16/2024]
Abstract
Brown adipose tissue (BAT) is a heater organ that expresses thermogenic uncoupling protein 1 (UCP1) to maintain high body temperatures during cold stress. BAT thermogenesis is considered an overarching mammalian trait, but its evolutionary origin is unknown. We show that adipose tissue of marsupials, which diverged from eutherian mammals ~150 million years ago, expresses a nonthermogenic UCP1 variant governed by a partial transcriptomic BAT signature similar to that found in eutherian beige adipose tissue. We found that the reconstructed UCP1 sequence of the common eutherian ancestor displayed typical thermogenic activity, whereas therian ancestor UCP1 is nonthermogenic. Thus, mammalian adipose tissue thermogenesis may have evolved in two distinct stages, with a prethermogenic stage in the common therian ancestor linking UCP1 expression to adipose tissue and thermal stress. We propose that in a second stage, UCP1 acquired its thermogenic function specifically in eutherians, such that the onset of mammalian BAT thermogenesis occurred only after the divergence from marsupials.
Collapse
Affiliation(s)
- Susanne Keipert
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Michael J Gaudry
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Maria Kutschke
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Michaela Keuper
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Margeoux A S Dela Rosa
- Biomedical Research Centre, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Yiming Cheng
- Institute of Neuronal Cell Biology, Technical University of Munich, 80802 Munich, Germany
- Munich Cluster of Systems Neurology, 81377 Munich, Germany
- Institute for Diabetes and Obesity (IDO), Helmholtz Zentrum München, 85764 Munich, Germany
| | - José M Monroy Kuhn
- Computational Discovery Research, Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC), Helmholtz Zentrum Munich, German Research Center for Environmental Health, German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Rutger Laterveer
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Camila A Cotrim
- Biomedical Research Centre, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Peter Giere
- Museum für Naturkunde-Leibniz Institute for Evolution and Biodiversity Science, 10115 Berlin, Germany
| | - Fabiana Perocchi
- Institute of Neuronal Cell Biology, Technical University of Munich, 80802 Munich, Germany
- Munich Cluster of Systems Neurology, 81377 Munich, Germany
- Institute for Diabetes and Obesity (IDO), Helmholtz Zentrum München, 85764 Munich, Germany
| | - Regina Feederle
- Monoclonal Antibody Core Facility, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Paul G Crichton
- Biomedical Research Centre, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | - Dominik Lutter
- Computational Discovery Research, Institute for Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC), Helmholtz Zentrum Munich, German Research Center for Environmental Health, German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Martin Jastroch
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| |
Collapse
|
28
|
Sharma AK, Khandelwal R, Wolfrum C. Futile cycles: Emerging utility from apparent futility. Cell Metab 2024; 36:1184-1203. [PMID: 38565147 DOI: 10.1016/j.cmet.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/15/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
Futile cycles are biological phenomena where two opposing biochemical reactions run simultaneously, resulting in a net energy loss without appreciable productivity. Such a state was presumed to be a biological aberration and thus deemed an energy-wasting "futile" cycle. However, multiple pieces of evidence suggest that biological utilities emerge from futile cycles. A few established functions of futile cycles are to control metabolic sensitivity, modulate energy homeostasis, and drive adaptive thermogenesis. Yet, the physiological regulation, implication, and pathological relevance of most futile cycles remain poorly studied. In this review, we highlight the abundance and versatility of futile cycles and propose a classification scheme. We further discuss the energetic implications of various futile cycles and their impact on basal metabolic rate, their bona fide and tentative pathophysiological implications, and putative drug interactions.
Collapse
Affiliation(s)
- Anand Kumar Sharma
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| | - Radhika Khandelwal
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Christian Wolfrum
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|
29
|
Tek NA, Şentüre ŞA, Ersoy N. Is Propolis a Potential Anti-Obesogenic Agent for Obesity? Curr Nutr Rep 2024; 13:186-193. [PMID: 38436884 PMCID: PMC11133030 DOI: 10.1007/s13668-024-00524-0] [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] [Accepted: 02/22/2024] [Indexed: 03/05/2024]
Abstract
PURPOSE OF REVIEW Propolis is a bee product that has been used for thousands of years. The chemical composition and biological activity of propolis, which has been investigated in the twentieth century, may vary according to location. Propolis polyphenols can induce thermogenesis in brown and beige fat tissue via the uncoupled protein-1 and creatinine kinase metabolic pathways. This review provides a comprehensive investigation of the structural and biological properties of propolis and provides insights into their promising potential strategies in body weight management. RECENT FINDINGS By raising overall energy expenditure, it might lead to body weight management. Furthermore, the phenolic components artepillin C, quercetin, catechin, and chlorogenic acid found in its composition may have anti-obesogenic effect by stimulating the sympathetic nervous system, enhancing browning in white adipose tissue, and triggering AMP-activated protein kinase activation and mitochondrial biogenesis. Propolis, a natural product, is effective in preventing obesity which is a contemporary pandemic.
Collapse
Affiliation(s)
- Nilüfer Acar Tek
- Faculty of Health Science, Department of Nutrition and Dietetic, Gazi University, Emek, Bişkek Main St. 6. St No: 2, 06490, Çankaya, Ankara, Turkey
| | - Şerife Akpınar Şentüre
- Faculty of Health Science, Department of Nutrition and Dietetic, Gazi University, Emek, Bişkek Main St. 6. St No: 2, 06490, Çankaya, Ankara, Turkey.
| | - Nursena Ersoy
- Faculty of Health Science, Department of Nutrition and Dietetic, Ankara University, Fatih Caddesi No:197/7 PK:06290, Keçiören, Ankara, Turkey
| |
Collapse
|
30
|
Liu L, Shang X, Ma L, Yan D, Adetula AA, Bai Y, Dong X. Transcriptomic Analyses Reveal the Effects of Walnut Kernel Cake on Adipose Deposition in Pigs. Genes (Basel) 2024; 15:667. [PMID: 38927603 PMCID: PMC11202485 DOI: 10.3390/genes15060667] [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: 04/03/2024] [Revised: 05/12/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
With the rising cost of animal feed protein, finding affordable and effective substitutes is crucial. Walnut kernel cake, a polyphenol-, fiber-, protein- and fat-rich byproduct of walnut oil extraction, has been underexplored as a potential protein replacement in pig feed. In this study, we found that feeding large Diqing Tibetan pigs walnut kernel cake promoted adipose deposition and improved pork quality during pig growth. Transcriptome analysis revealed the upregulation of genes ANGPTL8, CCNP, ETV4, and TRIB3, associated with adipose deposition. Pathway analysis highlighted enrichment in adipose deposition-related pathways, including PPAR, insulin, PI3K-Akt, Wnt, and MAPK signaling. Further analysis identified DEGs (differentially expressed genes) positively correlated with adipose-related traits, such as PER2 and PTGES. Single-cell transcriptome data pointed to the specific expression of CD248 and PTGES in adipocyte progenitor/stem cells (APSCs), pivotal for adipocyte differentiation and adipose deposition regulation. This study demonstrates walnut kernel cake's potential to substitute soybean cake in pig feed, providing high-quality protein and promoting adipose deposition. It offers insights into feed protein replacement, human functional food, fat metabolism, and related diseases, with marker genes and pathways supporting pig breeding and pork quality improvement.
Collapse
Affiliation(s)
- Lei Liu
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (L.L.); (D.Y.)
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China
| | - Xiaodan Shang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan 056038, China;
| | - Li Ma
- Department of Animal Husbandry and Veterinary Medicine, Yunnan Vocational and Technical College of Agriculture, Kunming 650212, China;
| | - Dawei Yan
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (L.L.); (D.Y.)
| | - Adeyinka Abiola Adetula
- Reproductive Biotechnology, Department of Molecular Life Sciences, TUM School of Life Sciences, Technical University Munich, 85354 Freising, Germany;
| | - Ying Bai
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan 056038, China;
| | - Xinxing Dong
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (L.L.); (D.Y.)
| |
Collapse
|
31
|
Ma L, Gilani A, Rubio-Navarro A, Cortada E, Li A, Reilly SM, Tang L, Lo JC. Adipsin and adipocyte-derived C3aR1 regulate thermogenic fat in a sex-dependent fashion. JCI Insight 2024; 9:e178925. [PMID: 38713526 PMCID: PMC11382875 DOI: 10.1172/jci.insight.178925] [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: 12/27/2023] [Accepted: 04/26/2024] [Indexed: 05/09/2024] Open
Abstract
Thermogenesis in beige/brown adipose tissues can be leveraged to combat metabolic disorders such as type 2 diabetes and obesity. The complement system plays pleiotropic roles in metabolic homeostasis and organismal energy balance with canonical effects on immune cells and noncanonical effects on nonimmune cells. The adipsin/C3a/C3a receptor 1 (C3aR1) pathway stimulates insulin secretion and sustains pancreatic β cell mass. However, its role in adipose thermogenesis has not been defined. Here, we show that male Adipsin/Cfd-knockout mice exhibited increased energy expenditure and white adipose tissue (WAT) browning. In addition, male adipocyte-specific C3aR1-knockout mice exhibited enhanced WAT thermogenesis and increased respiration. In stark contrast, female adipocyte-specific C3aR1-knockout mice displayed decreased brown fat thermogenesis and were cold intolerant. Female mice expressed lower levels of Adipsin in thermogenic adipocytes and adipose tissues than males. C3aR1 was also lower in female subcutaneous adipose tissue than in males. Collectively, these results reveal sexual dimorphism in the adipsin/C3a/C3aR1 axis in regulating adipose thermogenesis and defense against cold stress. Our findings establish a potentially new role of the alternative complement pathway in adaptive thermogenesis and highlight sex-specific considerations in potential therapeutic targets for metabolic diseases.
Collapse
Affiliation(s)
- Lunkun Ma
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Ankit Gilani
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Alfonso Rubio-Navarro
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Eric Cortada
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Ang Li
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Shannon M Reilly
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - James C Lo
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
32
|
Granath-Panelo M, Kajimura S. Mitochondrial heterogeneity and adaptations to cellular needs. Nat Cell Biol 2024; 26:674-686. [PMID: 38755301 DOI: 10.1038/s41556-024-01410-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/21/2024] [Indexed: 05/18/2024]
Abstract
Although it is well described that mitochondria are at the epicentre of the energy demands of a cell, it is becoming important to consider how each cell tailors its mitochondrial composition and functions to suit its particular needs beyond ATP production. Here we provide insight into mitochondrial heterogeneity throughout development as well as in tissues with specific energy demands and discuss how mitochondrial malleability contributes to cell fate determination and tissue remodelling.
Collapse
Affiliation(s)
- Melia Granath-Panelo
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, USA.
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Shingo Kajimura
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School and Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
33
|
Sharma AK, Khandelwal R, Wolfrum C. Futile lipid cycling: from biochemistry to physiology. Nat Metab 2024; 6:808-824. [PMID: 38459186 DOI: 10.1038/s42255-024-01003-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/02/2024] [Indexed: 03/10/2024]
Abstract
In the healthy state, the fat stored in our body isn't just inert. Rather, it is dynamically mobilized to maintain an adequate concentration of fatty acids (FAs) in our bloodstream. Our body tends to produce excess FAs to ensure that the FA availability is not limiting. The surplus FAs are actively re-esterified into glycerides, initiating a cycle of breakdown and resynthesis of glycerides. This cycle consumes energy without generating a new product and is commonly referred to as the 'futile lipid cycle' or the glyceride/FA cycle. Contrary to the notion that it's a wasteful process, it turns out this cycle is crucial for systemic metabolic homeostasis. It acts as a control point in intra-adipocyte and inter-organ cross-talk, a metabolic rheostat, an energy sensor and a lipid diversifying mechanism. In this Review, we discuss the metabolic regulation and physiological implications of the glyceride/FA cycle and its mechanistic underpinnings.
Collapse
Affiliation(s)
- Anand Kumar Sharma
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| | - Radhika Khandelwal
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Christian Wolfrum
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|
34
|
Su Y, Li X, Zhao J, Ji B, Zhao X, Feng J, Zhao J. Guanidinoacetic acid ameliorates hepatic steatosis and inflammation and promotes white adipose tissue browning in middle-aged mice with high-fat-diet-induced obesity. Food Funct 2024; 15:4515-4526. [PMID: 38567805 DOI: 10.1039/d3fo05201j] [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: 04/23/2024]
Abstract
Guanidinoacetic acid (GAA) is a naturally occurring amino acid derivative that plays a critical role in energy metabolism. In recent years, a growing body of evidence has emerged supporting the importance of GAA in metabolic dysfunction. Hence, we aimed to investigate the effects of GAA on hepatic and adipose tissue metabolism, as well as systemic inflammatory responses in obese middle-aged mice models and attempted to explore the underlying mechanism. We found that dietary supplementation of GAA inhibited inguinal white adipose tissue (iWAT) hypertrophy in high-fat diet (HFD)-fed mice. In addition, GAA supplementation observably decreased the levels of some systemic inflammatory factors, including IL-4, TNF-α, IL-1β, and IL-6. Intriguingly, GAA supplementation ameliorated hepatic steatosis and lipid deposition in HFD-fed mice, which was revealed by decreased levels of TG, TC, LDL-C, PPARγ, SREBP-1c, FASN, ACC, FABP1, and APOB and increased levels of HDL-C in the liver. Moreover, GAA supplementation increased the expression of browning markers and mitochondrial-related genes in the iWAT. Further investigation showed that dietary GAA promoted the browning of the iWAT via activating the AMPK/Sirt1 signaling pathway and might be associated with futile creatine cycling in obese mice. These results indicate that GAA has the potential to be used as an effective ingredient in dietary interventions and thus may play an important role in ameliorating and preventing HFD-induced obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Yuan Su
- College of Animal Sciences, Shanxi Agricultural University, Taigu 030801, PR China.
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Shanxi Agricultural University, Taigu 030801, PR China
| | - Xinrui Li
- College of Animal Sciences, Shanxi Agricultural University, Taigu 030801, PR China.
| | - Jiamin Zhao
- College of Animal Sciences, Shanxi Agricultural University, Taigu 030801, PR China.
| | - Bingzhen Ji
- College of Animal Sciences, Shanxi Agricultural University, Taigu 030801, PR China.
| | - Xiaoyi Zhao
- College of Animal Sciences, Shanxi Agricultural University, Taigu 030801, PR China.
| | - Jinxin Feng
- College of Animal Sciences, Shanxi Agricultural University, Taigu 030801, PR China.
| | - Junxing Zhao
- College of Animal Sciences, Shanxi Agricultural University, Taigu 030801, PR China.
- Shanxi Key Laboratory of Animal Genetics Resource Utilization and Breeding, Shanxi Agricultural University, Taigu 030801, PR China
| |
Collapse
|
35
|
Song H, Ren J, Yang L, Sun H, Yan G, Han Y, Wang X. Elucidation for the pharmacological effects and mechanism of Shen Bai formula in treating myocardial injury based on energy metabolism and serum metabolomic approaches. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117670. [PMID: 38160867 DOI: 10.1016/j.jep.2023.117670] [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: 09/23/2023] [Revised: 12/24/2023] [Accepted: 12/25/2023] [Indexed: 01/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shen Bai formula (SBF) is a proven effective traditional Chinese medicine for treating viral myocarditis (VMC) sequelae in clinic, and myocardial injury is the pathological basis of VMC sequelae. However, the pharmacological action and mechanism of SBF have not been systematically elucidated. AIM OF THE STUDY In present research, the doxorubicin-induced myocardial injury rat model was used to evaluate the efficacy of SBF, and energy metabolism and metabolomics approaches were applied to elucidate the effects of SBF on myocardial injury. MATERIALS AND METHODS Through energy metabolism measurement system and UPLC-Q-TOF-MS/MS oriented blood metabolomics, directly reflected the therapeutic effect of SBF at a macro level, and identified biomarkers of myocardial injury in microcosmic, revealing its metabolomic mechanism. RESULTS Results showed that SBF significantly improved the electrocardiogram (ECG), heart rate (HR), extent of myocardial tissue lesion, and ratio of heart and spleen. In addition, the serum levels of AST, CK, LDH, α-HBDH, cTnI, BNP, and MDA decreased, whereas SOD and ATP activity and content increased. Moreover, SBF increased locomotor activity and basic daily metabolism in rats with myocardial injury, restoring their usual level of energy metabolism. A total of 45 potential metabolomic biomarkers were identified. Among them, 44 biomarkers were significantly recalled by SBF, including representative biomarkers arachidonic acid (AA), 12-HETE, prostaglandin J2 (PGJ2), 15-deoxy-Δ-12,14-PGJ2, 15-keto-PGE2, 15(S)-HPETE, 15(S)-HETE, 8,11,14-eicosatrienoic acid and 9(S)-HODE, which involved AA metabolism, biosynthesis of unsaturated fatty acids and linoleic acid metabolism. CONCLUSION We successfully replicated a myocardial injury rat model with the intraperitoneal injection of doxorubicin, and elucidated the mechanism of SBF in treating myocardial injury. This key mechanism may be achieved by targeting action on COX, Alox, CYP, and 15-PGDH to increase or decrease the level of myocardial injury biomarker, and then emphatically interven in AA metabolism, biosynthesis of unsaturated fatty acids and linoleic acid metabolism, and participate in regulating purine metabolism, sphingolipid metabolism, primary bile acid biosynthesis, and steroid hormone synthesis.
Collapse
Affiliation(s)
- Hongwei Song
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Junling Ren
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau
| | - Le Yang
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China
| | - Hui Sun
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
| | - Guangli Yan
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Ying Han
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Xijun Wang
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau; State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China.
| |
Collapse
|
36
|
Zhao L, Yang H, Li M, Xiao M, Li X, Cheng L, Cheng W, Chen M, Zhao Y. Global gene expression profiling of perirenal brown adipose tissue whitening in goat kids reveals novel genes linked to adipose remodeling. J Anim Sci Biotechnol 2024; 15:47. [PMID: 38481287 PMCID: PMC10938744 DOI: 10.1186/s40104-024-00994-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/07/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Brown adipose tissue (BAT) is known to be capable of non-shivering thermogenesis under cold stimulation, which is related to the mortality of animals. In the previous study, we observed that goat BAT is mainly located around the kidney at birth, and changes to white adipose tissue (WAT) in the perirenal adipose tissue of goats within one month after birth. However, the regulatory factors underlying this change is remain unclear. In this study, we systematically studied the perirenal adipose tissue of goat kids in histological, cytological, and accompanying molecular level changes from 0 to 28 d after birth. RESULTS Our study found a higher mortality rate in winter-born goat kids, with goat birthing data statistics. Then we used thermal imaging revealing high temperature in goat hips at postnatal 0 d and gradually decrease during 28 d. This is consistent with the region of perirenal BAT deposition and highlights its critical role in energy expenditure and body temperature regulation in goat kids. Additionally, we found a series of changes of BAT during the first 28 d after birth, such as whitening, larger lipid droplets, decreased mitochondrial numbers, and down-regulation of key thermogenesis-related genes (UCP1, DIO2, UCP2, CIDEA, PPARGC1a, C/EBPb, and C/EBPa). Then, we used RNA-seq found specific marker genes for goat adipose tissue and identified 12 new marker genes for BAT and 10 new marker genes for WAT of goats. Furthermore, 12 candidate genes were found to potentially regulate goat BAT thermogenesis. The mechanism of the change of this biological phenomenon does not involve a large-scale death of brown adipocytes and subsequent proliferation of white adipocytes. While apoptosis may play a limited role, it is largely not critical in this transition process. CONCLUSIONS We concluded that perirenal BAT plays a crucial role in thermoregulation in newborn goat kids, with notable species differences in the expression of adipose tissue marker genes, and we highlighted some potential marker genes for goat BAT and WAT. Additionally, the change from BAT to WAT does not involve a large-scale death of brown adipocytes and subsequent proliferation of white adipocytes.
Collapse
Affiliation(s)
- Le Zhao
- College of Animal Science and Technology, Southwest University, Chongqing Key Laboratory of Herbivore Science, Chongqing, 400715, China
| | - Haili Yang
- College of Animal Science and Technology, Southwest University, Chongqing Key Laboratory of Herbivore Science, Chongqing, 400715, China
| | - Minhao Li
- College of Animal Science and Technology, Southwest University, Chongqing Key Laboratory of Herbivore Science, Chongqing, 400715, China
| | - Min Xiao
- College of Animal Science and Technology, Southwest University, Chongqing Key Laboratory of Herbivore Science, Chongqing, 400715, China
| | - Xingchun Li
- College of Animal Science and Technology, Southwest University, Chongqing Key Laboratory of Herbivore Science, Chongqing, 400715, China
| | - Lei Cheng
- College of Animal Science and Technology, Southwest University, Chongqing Key Laboratory of Herbivore Science, Chongqing, 400715, China
| | - Wenqiang Cheng
- College of Animal Science and Technology, Southwest University, Chongqing Key Laboratory of Herbivore Science, Chongqing, 400715, China
| | - Meixi Chen
- College of Animal Science and Technology, Southwest University, Chongqing Key Laboratory of Herbivore Science, Chongqing, 400715, China
| | - Yongju Zhao
- College of Animal Science and Technology, Southwest University, Chongqing Key Laboratory of Herbivore Science, Chongqing, 400715, China.
| |
Collapse
|
37
|
Dong M, Cheng Z, Jin W. UCP1 and CKB are parallel players in BAT. Cell Metab 2024; 36:459-460. [PMID: 38447527 DOI: 10.1016/j.cmet.2024.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 03/08/2024]
Abstract
It is generally believed that the contributions of the UCP1-independent thermogenic pathways are secondary to UCP1-mediated thermogenesis in BAT. Now, Rahbani et al. demonstrate in vivo that adaptive thermogenesis in brown adipose tissue is regulated by UCP1 and CKB in parallel.
Collapse
Affiliation(s)
- Meng Dong
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, 1 West Beichen Road No. 5, Chaoyang District, Beijing 100101, China
| | - Ziyu Cheng
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, 1 West Beichen Road No. 5, Chaoyang District, Beijing 100101, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Wanzhu Jin
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, 1 West Beichen Road No. 5, Chaoyang District, Beijing 100101, China; University of the Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
38
|
Rahbani JF, Bunk J, Lagarde D, Samborska B, Roesler A, Xiao H, Shaw A, Kaiser Z, Braun JL, Geromella MS, Fajardo VA, Koza RA, Kazak L. Parallel control of cold-triggered adipocyte thermogenesis by UCP1 and CKB. Cell Metab 2024; 36:526-540.e7. [PMID: 38272036 DOI: 10.1016/j.cmet.2024.01.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/27/2023] [Accepted: 01/02/2024] [Indexed: 01/27/2024]
Abstract
That uncoupling protein 1 (UCP1) is the sole mediator of adipocyte thermogenesis is a conventional viewpoint that has primarily been inferred from the attenuation of the thermogenic output of mice genetically lacking Ucp1 from birth (germline Ucp1-/-). However, germline Ucp1-/- mice harbor secondary changes within brown adipose tissue. To mitigate these potentially confounding ancillary changes, we constructed mice with inducible adipocyte-selective Ucp1 disruption. We find that, although germline Ucp1-/- mice succumb to cold-induced hypothermia with complete penetrance, most mice with the inducible deletion of Ucp1 maintain homeothermy in the cold. However, inducible adipocyte-selective co-deletion of Ucp1 and creatine kinase b (Ckb, an effector of UCP1-independent thermogenesis) exacerbates cold intolerance. Following UCP1 deletion or UCP1/CKB co-deletion from mature adipocytes, moderate cold exposure triggers the regeneration of mature brown adipocytes that coordinately restore UCP1 and CKB expression. Our findings suggest that thermogenic adipocytes utilize non-paralogous protein redundancy-through UCP1 and CKB-to promote cold-induced energy dissipation.
Collapse
Affiliation(s)
- Janane F Rahbani
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Jakub Bunk
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Damien Lagarde
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Bozena Samborska
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Anna Roesler
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Haopeng Xiao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Abhirup Shaw
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Zafir Kaiser
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Jessica L Braun
- Department of Kinesiology, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Mia S Geromella
- Department of Kinesiology, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Val A Fajardo
- Department of Kinesiology, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Robert A Koza
- MaineHealth Institute for Research, Scarborough, ME 04074, USA
| | - Lawrence Kazak
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada.
| |
Collapse
|
39
|
Park I, Kim KE, Kim J, Kim AK, Bae S, Jung M, Choi J, Mishra PK, Kim TM, Kwak C, Kang MG, Yoo CM, Mun JY, Liu KH, Lee KS, Kim JS, Suh JM, Rhee HW. Mitochondrial matrix RTN4IP1/OPA10 is an oxidoreductase for coenzyme Q synthesis. Nat Chem Biol 2024; 20:221-233. [PMID: 37884807 PMCID: PMC10830421 DOI: 10.1038/s41589-023-01452-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 09/17/2023] [Indexed: 10/28/2023]
Abstract
Targeting proximity-labeling enzymes to specific cellular locations is a viable strategy for profiling subcellular proteomes. Here, we generated transgenic mice (MAX-Tg) expressing a mitochondrial matrix-targeted ascorbate peroxidase. Comparative analysis of matrix proteomes from the muscle tissues showed differential enrichment of mitochondrial proteins. We found that reticulon 4-interacting protein 1 (RTN4IP1), also known as optic atrophy-10, is enriched in the mitochondrial matrix of muscle tissues and is an NADPH oxidoreductase. Interactome analysis and in vitro enzymatic assays revealed an essential role for RTN4IP1 in coenzyme Q (CoQ) biosynthesis by regulating the O-methylation activity of COQ3. Rtn4ip1-knockout myoblasts had markedly decreased CoQ9 levels and impaired cellular respiration. Furthermore, muscle-specific knockdown of dRtn4ip1 in flies resulted in impaired muscle function, which was reversed by dietary supplementation with soluble CoQ. Collectively, these results demonstrate that RTN4IP1 is a mitochondrial NAD(P)H oxidoreductase essential for supporting mitochondrial respiration activity in the muscle tissue.
Collapse
Affiliation(s)
- Isaac Park
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Kwang-Eun Kim
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Jeesoo Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
- Center for RNA Research, Institute for Basic Science, Seoul, Republic of Korea
| | - Ae-Kyeong Kim
- Metabolism and Neurophysiology Research Group, KRIBB, Daejeon, Republic of Korea
| | - Subin Bae
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Korea
| | - Minkyo Jung
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Jinhyuk Choi
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | | | - Taek-Min Kim
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Chulhwan Kwak
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Myeong-Gyun Kang
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Chang-Mo Yoo
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Ji Young Mun
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Kwang-Hyeon Liu
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Korea
| | - Kyu-Sun Lee
- Metabolism and Neurophysiology Research Group, KRIBB, Daejeon, Republic of Korea.
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea.
| | - Jong-Seo Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
- Center for RNA Research, Institute for Basic Science, Seoul, Republic of Korea.
| | - Jae Myoung Suh
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea.
| | - Hyun-Woo Rhee
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea.
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
40
|
Emanuelsson EB, Arif M, Reitzner SM, Perez S, Lindholm ME, Mardinoglu A, Daub C, Sundberg CJ, Chapman MA. Remodeling of the human skeletal muscle proteome found after long-term endurance training but not after strength training. iScience 2024; 27:108638. [PMID: 38213622 PMCID: PMC10783619 DOI: 10.1016/j.isci.2023.108638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/09/2023] [Accepted: 12/01/2023] [Indexed: 01/13/2024] Open
Abstract
Exercise training has tremendous systemic tissue-specific health benefits, but the molecular adaptations to long-term exercise training are not completely understood. We investigated the skeletal muscle proteome of highly endurance-trained, strength-trained, and untrained individuals and performed exercise- and sex-specific analyses. Of the 6,000+ proteins identified, >650 were differentially expressed in endurance-trained individuals compared with controls. Strikingly, 92% of the shared proteins with higher expression in both the male and female endurance groups were known mitochondrial. In contrast to the findings in endurance-trained individuals, minimal differences were found in strength-trained individuals and between females and males. Lastly, a co-expression network and comparative literature analysis revealed key proteins and pathways related to the health benefits of exercise, which were primarily related to differences in mitochondrial proteins. This network is available as an interactive database resource where investigators can correlate clinical data with global gene and protein expression data for hypothesis generation.
Collapse
Affiliation(s)
- Eric B. Emanuelsson
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Muhammad Arif
- Science for Life Laboratory, KTH – Royal Institute of Technology, 171 77 Stockholm, Sweden
| | - Stefan M. Reitzner
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Women’s and Children’s Health, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Sean Perez
- Department of Biology, Pomona College, Claremont, CA 91711, USA
| | - Maléne E. Lindholm
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH – Royal Institute of Technology, 171 77 Stockholm, Sweden
- Centre for Host–Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London WC2R 2LS, UK
| | - Carsten Daub
- Department of Biosciences and Nutrition, Karolinska Institutet, 171 77 Stockholm, Sweden
- Science for Life Laboratory, 171 65 Solna, Sweden
| | - Carl Johan Sundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, 141 52 Huddinge, Sweden
- Department of Learning, Informatics, Management and Ethics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Mark A. Chapman
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Integrated Engineering, University of San Diego, San Diego, CA 92110, USA
| |
Collapse
|
41
|
Eberhard C, Mosher EP, Bumpus N, Orsburn BC. Tenofovir Activation Is Diminished in the Brain and Liver of Creatine Kinase Brain-Type Knockout Mice. ACS Pharmacol Transl Sci 2024; 7:222-235. [PMID: 38230280 PMCID: PMC10789144 DOI: 10.1021/acsptsci.3c00250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/06/2023] [Accepted: 12/18/2023] [Indexed: 01/18/2024]
Abstract
Tenofovir (TFV) is a nucleotide reverse transcriptase inhibitor prescribed for the treatment and prevention of human immunodeficiency virus infection and the treatment of chronic hepatitis B virus infection. Here, we demonstrate that creatine kinase brain-type (CKB) can form tenofovir-diphosphate (TFV-DP), the pharmacologically active metabolite, in vitro and identify nine missense mutations (C74S, R96P, S128R, R132H, R172P, R236Q, C283S, R292Q, and H296R) that diminish this activity. Additional characterization of these mutations reveals that five (R96P, R132H, R236Q, C283S, and R292Q) have ATP dephosphorylation catalytic efficiencies less than 20% of those of the wild type (WT), and seven (C74S, R96P, R132H, R172P, R236Q, C283S, and H296P) induce thermal instabilities. To determine the extent CKB contributes to TFV activation in vivo, we generated a CKB knockout mouse strain, Ckbtm1Nnb. Using an in vitro assay, we show that brain lysates of Ckbtm1Nnb male and female mice form 70.5 and 77.4% less TFV-DP than wild-type brain lysates of the same sex, respectively. Additionally, we observe that Ckbtm1Nnb male mice treated with tenofovir disoproxil fumarate for 14 days exhibit a 22.8% reduction in TFV activation in the liver compared to wild-type male mice. Lastly, we utilize mass spectrometry-based proteomics to elucidate the impact of the knockout on the abundance of nucleotide and small molecule kinases in the brain and liver, adding to our understanding of how the loss of CKB may be impacting tenofovir activation in these tissues. Together, our data suggest that disruptions in CKB may lower levels of active drugs in the brain and liver.
Collapse
Affiliation(s)
- Colten
D. Eberhard
- Department of Pharmacology
and Molecular Sciences, Johns Hopkins University
School of Medicine, Baltimore, Maryland 21205, United States
| | - Eric P. Mosher
- Department of Pharmacology
and Molecular Sciences, Johns Hopkins University
School of Medicine, Baltimore, Maryland 21205, United States
| | - Namandjé
N. Bumpus
- Department of Pharmacology
and Molecular Sciences, Johns Hopkins University
School of Medicine, Baltimore, Maryland 21205, United States
| | - Benjamin C. Orsburn
- Department of Pharmacology
and Molecular Sciences, Johns Hopkins University
School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
42
|
Engin A. Reappraisal of Adipose Tissue Inflammation in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:297-327. [PMID: 39287856 DOI: 10.1007/978-3-031-63657-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Chronic low-grade inflammation is a central component in the pathogenesis of obesity-related expansion of adipose tissue and complications in other metabolic tissues. Five different signaling pathways are defined as dominant determinants of adipose tissue inflammation: These are increased circulating endotoxin due to dysregulation in the microbiota-gut-brain axis, systemic oxidative stress, macrophage accumulation, and adipocyte death. Finally, the nucleotide-binding and oligomerization domain (NOD) leucine-rich repeat family pyrin domain-containing 3 (NLRP3) inflammasome pathway is noted to be a key regulator of metabolic inflammation. The NLRP3 inflammasome and associated metabolic inflammation play an important role in the relationships among fatty acids and obesity. Several highly active molecules, including primarily leptin, resistin, adiponectin, visfatin, and classical cytokines, are abundantly released from adipocytes. The most important cytokines that are released by inflammatory cells infiltrating obese adipose tissue are tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), monocyte chemoattractant protein 1 (MCP-1) (CCL-2), and IL-1. All these molecules mentioned above act on immune cells, causing local and then general inflammation. Three metabolic pathways are noteworthy in the development of adipose tissue inflammation: toll-like receptor 4 (TLR4)/phosphatidylinositol-3'-kinase (PI3K)/Protein kinase B (Akt) signaling pathway, endoplasmic reticulum (ER) stress-derived unfolded protein response (UPR), and inhibitor of nuclear factor kappa-B kinase beta (IKKβ)-nuclear factor kappa B (NF-κB) pathway. In fact, adipose tissue inflammation is an adaptive response that contributes to a visceral depot barrier that effectively filters gut-derived endotoxin. Excessive fatty acid release worsens adipose tissue inflammation and contributes to insulin resistance. However, suppression of adipose inflammation in obesity with anti-inflammatory drugs is not a rational solution and paradoxically promotes insulin resistance, despite beneficial effects on weight gain. Inflammatory pathways in adipocytes are indeed indispensable for maintaining systemic insulin sensitivity. Cannabinoid type 1 receptor (CB1R) is important in obesity-induced pro-inflammatory response; however, blockade of CB1R, contrary to anti-inflammatory drugs, breaks the links between insulin resistance and adipose tissue inflammation. Obesity, however, could be decreased by improving leptin signaling, white adipose tissue browning, gut microbiota interactions, and alleviating inflammation. Furthermore, capsaicin synthesized by chilies is thought to be a new and promising therapeutic option in obesity, as it prevents metabolic endotoxemia and systemic chronic low-grade inflammation caused by high-fat diet.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
43
|
Allebrandt Neto EW, Rondon E Silva J, Santos SF, de França Lemes SA, Kawashita NH, Peron Pereira M. The futile creatine cycle and the synthesis of fatty acids in inguinal white adipose tissue from growing rats, submitted to a hypoprotein-hyperglycidic diet for 15 days. Lipids 2024; 59:3-12. [PMID: 38223990 DOI: 10.1002/lipd.12384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 11/30/2023] [Accepted: 01/02/2024] [Indexed: 01/16/2024]
Abstract
The low-protein, high-carbohydrate (LPHC) diet administered to growing rats soon after weaning, for 15 days, promoted an increase in energy expenditure by uncoupling protein 1 (UCP1) in interscapular brown adipose tissue, and also due to the occurrence of the browning process in the perirenal white adipose tissue (periWAT). However, we believe that inguinal white adipose tissue (ingWAT) may also contribute to energy expenditure through other mechanisms. Therefore, the aim of this work is to investigate the presence of the futile creatine cycle, and the origin of lipids in ingWAT, since that tissue showed an increase in the lipids content in rats submitted to the LPHC diet for 15 days. We observed increases in creatine kinase and alkaline phosphatase activity in ingWAT, of the LPHC animals. The mitochondrial Nicotinamide adenine dinucleotide reduced/nicotinamide adenine dinucleotide oxidized ratio is lower in ingWAT of LPHC animals. In the LPHC animals treated with β-guanidinopropionic acid, the extracellular uptake of creatine in ingWAT was lower, as was the rectal temperature. Regarding lipid metabolism, we observed that in ingWAT, lipolysis in vitro when stimulated with noradrenaline is lower, and there were no changes in baseline levels. In addition, increases in the activity of enzymes were also observed: malic, glucose-6-phosphate dehydrogenase, and ATP-citrate lyase, in addition to an increase in the PPARγ content. The results show the occurrence of the futile creatine cycle in ingWAT, and that the increase in the relative mass may be due to an increase in de novo fatty acid synthesis.
Collapse
Affiliation(s)
| | | | | | | | - Nair Honda Kawashita
- Department of Chemistry, Federal University of Mato Grosso, Cuiabá, Mato Grosso, Brazil
| | - Mayara Peron Pereira
- Department of Chemistry, Federal University of Mato Grosso, Cuiabá, Mato Grosso, Brazil
| |
Collapse
|
44
|
Terzioglu M, Veeroja K, Montonen T, Ihalainen TO, Salminen TS, Bénit P, Rustin P, Chang YT, Nagai T, Jacobs HT. Mitochondrial temperature homeostasis resists external metabolic stresses. eLife 2023; 12:RP89232. [PMID: 38079477 PMCID: PMC10712956 DOI: 10.7554/elife.89232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
Based on studies with a fluorescent reporter dye, Mito Thermo Yellow (MTY), and the genetically encoded gTEMP ratiometric fluorescent temperature indicator targeted to mitochondria, the temperature of active mitochondria in four mammalian and one insect cell line was estimated to be up to 15°C above that of the external environment to which the cells were exposed. High mitochondrial temperature was maintained in the face of a variety of metabolic stresses, including substrate starvation or modification, decreased ATP demand due to inhibition of cytosolic protein synthesis, inhibition of the mitochondrial adenine nucleotide transporter and, if an auxiliary pathway for electron transfer was available via the alternative oxidase, even respiratory poisons acting downstream of oxidative phosphorylation (OXPHOS) complex I. We propose that the high temperature of active mitochondria is an inescapable consequence of the biochemistry of OXPHOS and is homeostatically maintained as a primary feature of mitochondrial metabolism.
Collapse
Affiliation(s)
- Mügen Terzioglu
- Faculty of Medicine and Health Technology, Tampere UniversityTampereFinland
| | - Kristo Veeroja
- Faculty of Medicine and Health Technology, Tampere UniversityTampereFinland
| | - Toni Montonen
- Faculty of Medicine and Health Technology, Tampere UniversityTampereFinland
| | - Teemu O Ihalainen
- Faculty of Medicine and Health Technology, Tampere UniversityTampereFinland
| | - Tiina S Salminen
- Faculty of Medicine and Health Technology, Tampere UniversityTampereFinland
| | - Paule Bénit
- Université Paris Cité, Inserm, Maladies Neurodéveloppementales et NeurovasculairesParisFrance
| | - Pierre Rustin
- Université Paris Cité, Inserm, Maladies Neurodéveloppementales et NeurovasculairesParisFrance
| | - Young-Tae Chang
- SANKEN (The Institute of Scientific and Industrial Research), Osaka UniversityIbarakiJapan
| | | | - Howard T Jacobs
- Faculty of Medicine and Health Technology, Tampere UniversityTampereFinland
- Department of Environment and Genetics, La Trobe UniversityMelbourneAustralia
| |
Collapse
|
45
|
Duerre DJ, Hansen JK, John S, Jen A, Carrillo N, Bui H, Bao Y, Fabregat M, Overmeyer K, Shishkova E, Keller MP, Anderson RA, Cryns VL, Attie AD, Coon JJ, Fan J, Galmozzi A. Heme biosynthesis regulates BCAA catabolism and thermogenesis in brown adipose tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.28.568893. [PMID: 38076785 PMCID: PMC10705273 DOI: 10.1101/2023.11.28.568893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
With age, people tend to accumulate body fat and reduce energy expenditure 1 . Brown (BAT) and beige adipose tissue dissipate heat and increase energy expenditure via the activity of the uncoupling protein UCP1 and other thermogenic futile cycles 2,3 . The activity of brown and beige depots inversely correlates with BMI and age 4-11 , suggesting that promoting thermogenesis may be an effective approach for combating age-related metabolic disease 12-15 . Heme is an enzyme cofactor and signaling molecule that we recently showed to regulate BAT function 16 . Here, we show that heme biosynthesis is the primary contributor to intracellular heme levels in brown adipocytes. Inhibition of heme biosynthesis leads to mitochondrial dysfunction and reduction in UCP1. Although supplementing heme can restore mitochondrial function in heme-synthesis-deficient cells, the downregulation of UCP1 persists due to the accumulation of the heme precursors, particularly propionyl-CoA, which is a product of branched-chain amino acids (BCAA) catabolism. Cold exposure promotes BCAA uptake in BAT, and defects in BCAA catabolism in this tissue hinder thermogenesis 17 . However, BCAAs' contribution to the TCA cycle in BAT and WAT never exceeds 2% of total TCA flux 18 . Our work offers a way to integrate current literature by describing heme biosynthesis as an important metabolic sink for BCAAs.
Collapse
|
46
|
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
|
47
|
Fan Z, Jia W. Long Short-Term Memory-Based Multiomics Reveal Lactobacillus casei-Derived Postbiotics Inhibiting Lipids Digestion via Mediating the Upregulation of α-Helices in Lipase. Mol Nutr Food Res 2023; 67:e2300336. [PMID: 37753826 DOI: 10.1002/mnfr.202300336] [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: 05/24/2023] [Revised: 08/09/2023] [Indexed: 09/28/2023]
Abstract
SCOPE The antiobesity function of probiotics has been declared, while the application in high-risk patients and coding side effect has focused attention to postbiotics. This investigation profiles the mechanism of postbiotics affecting lipid digestion at molecular level, and establishes a momentous foundation for the clinical application of postbiotics in obesity suppression. METHODS AND RESULTS An operational framework for butter digestion is constructed to collect the digests in the intestine at 0, 40, 80, and 120 min with various postbiotics supplement. A total of 227 lipids and 414 metabolites are detected by pseudo-targeted lipidomics integrated with the long short-term memory-based metabolomics, and the triacylglycerol (TG, from 134.1 to 184.7 mg kg-1 ) and diacylglycerol (DG, from 4.2 to 8.4 mg kg-1 ) are identified as significantly different lipids with or without postbiotics supplement. A total of eight substances related to the inhibition of gastric lipase and pancreatic lipase are screened through the molecular simulation computation in silicon and enzymatic reaction kinetics, and thus curtailing the bioaccessibility of lipids. CONCLUSIONS Lactobacillus casei JCM1134-derived postbiotics propel the structure of lipase to aggregate by increasing the α-helix, and thus hampering the digestion of triglycerides through noncompetitive inhibition.
Collapse
Affiliation(s)
- Zibian Fan
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Wei Jia
- School of Food and Biological Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China
- Shaanxi Research Institute of Agricultural Products Processing Technology, Xi'an, 710021, China
| |
Collapse
|
48
|
El-Yazbi AF, Elrewiny MA, Habib HM, Eid AH, Elzahhar PA, Belal ASF. Thermogenic Modulation of Adipose Depots: A Perspective on Possible Therapeutic Intervention with Early Cardiorenal Complications of Metabolic Impairment. Mol Pharmacol 2023; 104:187-194. [PMID: 37567782 DOI: 10.1124/molpharm.123.000704] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Cardiovascular complications of diabetes and obesity remain a major cause for morbidity and mortality worldwide. Despite significant advances in the pharmacotherapy of metabolic disease, the available approaches do not prevent or slow the progression of complications. Moreover, a majority of patients present with significant vascular involvement at early stages of dysfunction prior to overt metabolic changes. The lack of disease-modifying therapies affects millions of patients globally, causing a massive economic burden due to these complications. Significantly, adipose tissue inflammation was implicated in the pathogenesis of metabolic syndrome, diabetes, and obesity. Specifically, perivascular adipose tissue (PVAT) and perirenal adipose tissue (PRAT) depots influence cardiovascular and renal structure and function. Accumulating evidence implicates localized PVAT/PRAT inflammation as the earliest response to metabolic impairment leading to cardiorenal dysfunction. Increased mitochondrial uncoupling protein 1 (UCP1) expression and function lead to PVAT/PRAT hypoxia and inflammation as well as vascular, cardiac, and renal dysfunction. As UCP1 function remains an undruggable target so far, modulation of the augmented UCP1-mediated PVAT/PRAT thermogenesis constitutes a lucrative target for drug development to mitigate early cardiorenal involvement. This can be achieved either by subtle targeted reduction in UCP-1 expression using innovative proteolysis activating chimeric molecules (PROTACs) or by supplementation with cyclocreatine phosphate, which augments the mitochondrial futile creatine cycling and thus decreases UCP1 activity, enhances the efficiency of oxygen use, and reduces hypoxia. Once developed, these molecules will be first-in-class therapeutic tools to directly interfere with and reverse the earliest pathology underlying cardiac, vascular, and renal dysfunction accompanying the early metabolic deterioration. SIGNIFICANCE STATEMENT: Adipose tissue dysfunction plays a major role in the pathogenesis of metabolic diseases and their complications. Although mitochondrial alterations are common in metabolic impairment, it was only recently shown that the early stages of metabolic challenge involve inflammatory changes in select adipose depots associated with increased uncoupling protein 1 thermogenesis and hypoxia. Manipulating this mode of thermogenesis can help mitigate the early inflammation and the consequent cardiorenal complications.
Collapse
Affiliation(s)
- Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology (A.F.E.-Y.) and Department of Pharmaceutical Chemistry (P.A.E., A.S.F.B.), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Research and Innovation Hub, Alamein International University, Alamein, Egypt (A.F.E.-Y., M.A.E., H.M.H.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Mohamed A Elrewiny
- Department of Pharmacology and Toxicology (A.F.E.-Y.) and Department of Pharmaceutical Chemistry (P.A.E., A.S.F.B.), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Research and Innovation Hub, Alamein International University, Alamein, Egypt (A.F.E.-Y., M.A.E., H.M.H.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Hosam M Habib
- Department of Pharmacology and Toxicology (A.F.E.-Y.) and Department of Pharmaceutical Chemistry (P.A.E., A.S.F.B.), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Research and Innovation Hub, Alamein International University, Alamein, Egypt (A.F.E.-Y., M.A.E., H.M.H.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Ali H Eid
- Department of Pharmacology and Toxicology (A.F.E.-Y.) and Department of Pharmaceutical Chemistry (P.A.E., A.S.F.B.), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Research and Innovation Hub, Alamein International University, Alamein, Egypt (A.F.E.-Y., M.A.E., H.M.H.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Perihan A Elzahhar
- Department of Pharmacology and Toxicology (A.F.E.-Y.) and Department of Pharmaceutical Chemistry (P.A.E., A.S.F.B.), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Research and Innovation Hub, Alamein International University, Alamein, Egypt (A.F.E.-Y., M.A.E., H.M.H.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Ahmed S F Belal
- Department of Pharmacology and Toxicology (A.F.E.-Y.) and Department of Pharmaceutical Chemistry (P.A.E., A.S.F.B.), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Research and Innovation Hub, Alamein International University, Alamein, Egypt (A.F.E.-Y., M.A.E., H.M.H.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| |
Collapse
|
49
|
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
|
50
|
Cai J, Wang F, Shao M. The Emerging Importance of Mitochondria in White Adipocytes: Neither Last nor Least. Endocrinol Metab (Seoul) 2023; 38:493-503. [PMID: 37816498 PMCID: PMC10613775 DOI: 10.3803/enm.2023.1813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/05/2023] [Accepted: 09/14/2023] [Indexed: 10/12/2023] Open
Abstract
The growing recognition of mitochondria's crucial role in the regulation of white adipose tissue remodeling and energy balance underscores its significance. The marked metabolic diversity of mitochondria provides the molecular and cellular foundation for enabling adipose tissue plasticity in response to various metabolic cues. Effective control of mitochondrial function at the cellular level, not only in thermogenic brown and beige adipocytes but also in energy-storing white adipocytes, exerts a profound influence on adipose homeostasis. Furthermore, mitochondria play a pivotal role in intercellular communication within adipose tissue via production of metabolites with signaling properties. A more comprehensive understanding of mitochondrial regulation within white adipocytes will empower the development of targeted and efficacious strategies to enhance adipose function, leading to advancements in overall metabolic health.
Collapse
Affiliation(s)
- Juan Cai
- CAS Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Fenfen Wang
- Department of Anesthesiology, Critical Care and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, TX, USA
| | - Mengle Shao
- CAS Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|