1
|
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
|
2
|
Haddish K, Yun JW. Silencing of dopamine receptor D5 inhibits the browning of 3T3-L1 adipocytes and ATP-consuming futile cycles in C2C12 muscle cells. Arch Physiol Biochem 2024; 130:555-567. [PMID: 37140438 DOI: 10.1080/13813455.2023.2206983] [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: 12/21/2022] [Accepted: 04/19/2023] [Indexed: 05/05/2023]
Abstract
BACKGROUND As a part of the catecholamines, dopamine receptors (DRs) have not been extensively studied like β3-AR in the thermogenesis process. The present study investigates the effect of DRD5 in browning events and ATP-consuming futile cycles. METHODS siRNA technology, qPCR, immunoblot analysis, immunofluorescence, and staining methods were used to investigate the effect of DRD5 on 3T3-L1 and C2C12 cells. RESULTS siDdr5 increased lipogenesis-associated effectors, and adipogenesis markers while reducing the expression of beige fat effectors. ATP-consuming futile cycle markers were also reduced following the siDrd5. On the contrary, pharmacological activation of DRD5 stimulated these effectors. Our mechanistic studies elucidated that DRD5 mediates fat browning via the cAMP-PKA-p38 MAPK signalling pathway in 3T3-L1 cells as well as the cAMP-SERCA-RyR pathway for the ATP-consuming futile cycles in both cells. CONCLUSIONS siDrd5 positively regulates browning and ATP-consuming futile cycles, and understanding its functions will provide insights into novel strategies to treat obesity.
Collapse
Affiliation(s)
- Kiros Haddish
- Department of Biotechnology, Daegu University, Gyeongbuk, Republic of Korea
| | - Jong Won Yun
- Department of Biotechnology, Daegu University, Gyeongbuk, Republic of Korea
| |
Collapse
|
3
|
Gonzalez DE, Forbes SC, Zapp A, Jagim A, Luedke J, Dickerson BL, Root A, Gil A, Johnson SE, Coles M, Brager A, Sowinski RJ, Candow DG, Kreider RB. Fueling the Firefighter and Tactical Athlete with Creatine: A Narrative Review of a Key Nutrient for Public Safety. Nutrients 2024; 16:3285. [PMID: 39408252 PMCID: PMC11478539 DOI: 10.3390/nu16193285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objectives: Firefighters, tactical police officers, and warriors often engage in periodic, intermittent, high-intensity physical work in austere environmental conditions and have a heightened risk of premature mortality. In addition, tough decision-making challenges, routine sleep deprivation, and trauma exacerbate this risk. Therefore, identifying strategies to bolster these personnel's health and occupational performance is critical. Creatine monohydrate (CrM) supplementation may offer several benefits to firefighters and tactical athletes (e.g., police, security, and soldiers) due to its efficacy regarding physical performance, muscle, cardiovascular health, mental health, and cognitive performance. Methods: We conducted a narrative review of the literature with a focus on the benefits and application of creatine monohydrate among firefighters. Results: Recent evidence demonstrates that CrM can improve anaerobic exercise capacity and muscular fitness performance outcomes and aid in thermoregulation, decision-making, sleep, recovery from traumatic brain injuries (TBIs), and mental health. Emerging evidence also suggests that CrM may confer an antioxidant/anti-inflammatory effect, which may be particularly important for firefighters and those performing tactical occupations exposed to oxidative and physiological stress, which can elicit systemic inflammation and increase the risk of chronic diseases. Conclusions: This narrative review highlights the potential applications of CrM for related tactical occupations, with a particular focus on firefighters, and calls for further research into these populations.
Collapse
Affiliation(s)
- Drew E. Gonzalez
- Exercise and Sport Nutrition Laboratory, Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX 77843, USA; (B.L.D.); (S.E.J.); (R.J.S.); (R.B.K.)
- Tactical Athlete Research Unit, Texas A&M University, College Station, TX 77843, USA;
| | - Scott C. Forbes
- Department of Physical Education Studies, Faculty of Education, Brandon University, Brandon, MB R7A 6A9, Canada;
| | | | - Andrew Jagim
- Sports Medicine, Mayo Clinic Health System, La Crosse, WI 54601, USA;
| | - Joel Luedke
- Olmsted Medical Center-Sports Medicine, La Crosse, WI 54601, USA;
| | - Broderick L. Dickerson
- Exercise and Sport Nutrition Laboratory, Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX 77843, USA; (B.L.D.); (S.E.J.); (R.J.S.); (R.B.K.)
| | | | - Adriana Gil
- College of Medicine, University of Houston, Houston, TX 77021, USA;
| | - Sarah E. Johnson
- Exercise and Sport Nutrition Laboratory, Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX 77843, USA; (B.L.D.); (S.E.J.); (R.J.S.); (R.B.K.)
- Tactical Athlete Research Unit, Texas A&M University, College Station, TX 77843, USA;
| | - Macilynn Coles
- Tactical Athlete Research Unit, Texas A&M University, College Station, TX 77843, USA;
| | - Allison Brager
- U.S. Army John F. Kennedy Special Warfare Center and School, Fort Liberty, NC 48397, USA;
| | - Ryan J. Sowinski
- Exercise and Sport Nutrition Laboratory, Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX 77843, USA; (B.L.D.); (S.E.J.); (R.J.S.); (R.B.K.)
| | - Darren G. Candow
- Faculty of Kinesiology and Health Studies, University of Regina, Regina, SK S4S 0A2, Canada;
| | - Richard B. Kreider
- Exercise and Sport Nutrition Laboratory, Department of Kinesiology and Sport Management, Texas A&M University, College Station, TX 77843, USA; (B.L.D.); (S.E.J.); (R.J.S.); (R.B.K.)
| |
Collapse
|
4
|
Yang J, Yuan M, Zhang W. The major biogenic amine metabolites in mood disorders. Front Psychiatry 2024; 15:1460631. [PMID: 39381610 PMCID: PMC11458445 DOI: 10.3389/fpsyt.2024.1460631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024] Open
Abstract
Mood disorders, including major depressive disorder and bipolar disorder, have a profound impact on more than 300 million people worldwide. It has been demonstrated mood disorders were closely associated with deviations in biogenic amine metabolites, which are involved in numerous critical physiological processes. The peripheral and central alteration of biogenic amine metabolites in patients may be one of the potential pathogeneses of mood disorders. This review provides a concise overview of the latest research on biogenic amine metabolites in mood disorders, such as histamine, kynurenine, and creatine. Further studies need larger sample sizes and multi-center collaboration. Investigating the changes of biogenic amine metabolites in mood disorders can provide biological foundation for diagnosis, offer guidance for more potent treatments, and aid in elucidating the biological mechanisms underlying mood disorders.
Collapse
Affiliation(s)
- Jingyi Yang
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Minlan Yuan
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Zhang
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
- Big Data Center, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Hankir MK. Creating a picture of brown fat with creatine-CEST. Trends Endocrinol Metab 2024:S1043-2760(24)00252-2. [PMID: 39256118 DOI: 10.1016/j.tem.2024.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/12/2024]
Abstract
Accurate assessment of brown fat thermogenesis by non-invasive means remains challenging. Writing in Nature Metabolism, Cai et al. leverage the futile creatine cycling characteristic of thermogenic adipocytes to show that a type of magnetic resonance imaging (MRI) technique sensitive to endogenous creatine levels faithfully tracks brown fat thermogenesis in rodents and in humans.
Collapse
Affiliation(s)
- Mohammed K Hankir
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
6
|
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] [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
|
7
|
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
|
8
|
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
|
9
|
Zhan H, Wang W, Ge Y, Liang Y, Wang J, Xu Y, Wu S, Peng L, He Z. trans-Palmitoleic acid promotes adipose thermogenesis to reduce obesity via hypothalamic FFAR1 signaling. Food Funct 2024; 15:4627-4641. [PMID: 38592736 DOI: 10.1039/d4fo00452c] [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: 04/10/2024]
Abstract
Diet-induced thermogenesis (DIT) is crucial for maintaining body weight homeostasis, and the role of dietary fatty acids in modulating DIT is essential. However, the underlying mechanism of fatty acid regulated diet-induced thermogenesis remains elusive. Utilizing the diet- and genetic ablation-induced obese mice models, we found that the C16 unsaturated fatty acids, trans-palmitoleic acid (TPA) and cis-palmitoleic acid (CPA), significantly increased the energy expenditure by promoting the thermogenesis of brown adipose tissues and the production of beige cells in white adipose. As a result, there is a significant reduction in the occurrence of obesity, associated hepatic steatosis and hyperglycemia. Notably, TPA exhibited more potent effects on promoting DIT and alleviating obesity than CPA did. Using inhibitor and gene deletion mice models, we unveiled that TPA acted as a signaling molecule to play a biological function, which could be sensed by the hypothalamic FFAR1 to activate the sympathetic nervous system in promoting adipose tissue thermogenesis. Together, these results demonstrate the underlying mechanism of free fatty acids associated-DIT and will provide fresh insights into the roles of trans-fatty acids in the development of obesity.
Collapse
Affiliation(s)
- Huidong Zhan
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China.
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wanjing Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Yueping Ge
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China.
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yixiao Liang
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China.
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jing Wang
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China.
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yang Xu
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China.
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shanshan Wu
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China.
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Li Peng
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China.
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhao He
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration and Practice Center, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China.
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| |
Collapse
|
10
|
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: 14] [Impact Index Per Article: 14.0] [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
|
11
|
Chavanne A, Jacobi D. Precision medicine in endocrinology: Unraveling metabolic health through time-restricted eating. ANNALES D'ENDOCRINOLOGIE 2024; 85:63-69. [PMID: 38101564 DOI: 10.1016/j.ando.2023.12.001] [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: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023]
Abstract
As a promising avenue in nutrition, intermittent fasting, particularly time-restricted eating like the 8/16 protocol, requires careful individualization. This approach involves voluntary food restriction interspersed with normal eating, aiming to align with inner circadian rhythms for potential benefits in metabolism and weight management. Endocrinologists, responding to patient interest and backed by evidence-based medicine, can now delve into the intricacies of time-restricted eating. They consider each patient's unique medical history and expectations, integrating this approach into tailored treatment plans in a personalized medicine approach. Ongoing research is essential to deepen our comprehension of how time-restricted eating influences metabolic health, enabling the development of precise recommendations suitable for diverse populations and various clinical conditions. While time-restricted eating is a relevant metabolic approach, endocrinologists should exercise caution to prevent the promotion of eating disorders due to its restrictive nature.
Collapse
Affiliation(s)
- Albane Chavanne
- CHU de Nantes, Nantes Université, CNRS, INSERM, l'Institut du thorax, Nantes, France
| | - David Jacobi
- Institut de recherche en santé de Nantes Université, 8, quai Moncousu, 44000 Nantes, France.
| |
Collapse
|
12
|
Rashidi A, Billingham LK, Zolp A, Chia TY, Silvers C, Katz JL, Park CH, Delay S, Boland L, Geng Y, Markwell SM, Dmello C, Arrieta VA, Zilinger K, Jacob IM, Lopez-Rosas A, Hou D, Castro B, Steffens AM, McCortney K, Walshon JP, Flowers MS, Lin H, Wang H, Zhao J, Sonabend A, Zhang P, Ahmed AU, Brat DJ, Heiland DH, Lee-Chang C, Lesniak MS, Chandel NS, Miska J. Myeloid cell-derived creatine in the hypoxic niche promotes glioblastoma growth. Cell Metab 2024; 36:62-77.e8. [PMID: 38134929 PMCID: PMC10842612 DOI: 10.1016/j.cmet.2023.11.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 05/08/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023]
Abstract
Glioblastoma (GBM) is a malignancy dominated by the infiltration of tumor-associated myeloid cells (TAMCs). Examination of TAMC metabolic phenotypes in mouse models and patients with GBM identified the de novo creatine metabolic pathway as a hallmark of TAMCs. Multi-omics analyses revealed that TAMCs surround the hypoxic peri-necrotic regions of GBM and express the creatine metabolic enzyme glycine amidinotransferase (GATM). Conversely, GBM cells located within these same regions are uniquely specific in expressing the creatine transporter (SLC6A8). We hypothesized that TAMCs provide creatine to tumors, promoting GBM progression. Isotopic tracing demonstrated that TAMC-secreted creatine is taken up by tumor cells. Creatine supplementation protected tumors from hypoxia-induced stress, which was abrogated with genetic ablation or pharmacologic inhibition of SLC6A8. Lastly, inhibition of creatine transport using the clinically relevant compound, RGX-202-01, blunted tumor growth and enhanced radiation therapy in vivo. This work highlights that myeloid-to-tumor transfer of creatine promotes tumor growth in the hypoxic niche.
Collapse
Affiliation(s)
- Aida Rashidi
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Leah K Billingham
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Andrew Zolp
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Tzu-Yi Chia
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Caylee Silvers
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Joshua L Katz
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Cheol H Park
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Suzi Delay
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Lauren Boland
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital, Chicago, IL, USA
| | - Yuheng Geng
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Steven M Markwell
- Department of Pathology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Crismita Dmello
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Victor A Arrieta
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Kaylee Zilinger
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Irene M Jacob
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Aurora Lopez-Rosas
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - David Hou
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Brandyn Castro
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Alicia M Steffens
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Kathleen McCortney
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Jordain P Walshon
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Mariah S Flowers
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Hanchen Lin
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Hanxiang Wang
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Junfei Zhao
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Adam Sonabend
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Peng Zhang
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Atique U Ahmed
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Daniel J Brat
- Department of Pathology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Dieter H Heiland
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA; Microenvironment and Immunology Research Laboratory, Medical Center, University of Freiburg, 79106 Freiburg, Germany; Department of Neurosurgery, Medical Center, University of Freiburg, 79106 Freiburg, Germany. German Cancer Consortium (DKTK), partner site Freiburg, Freiburg, Germany
| | - Catalina Lee-Chang
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Maciej S Lesniak
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Navdeep S Chandel
- Department of Medicine, Feinberg School of Medicine, Northwestern University, 676 North St. Clair Street, Suite 2330, Chicago, IL 60611, USA
| | - Jason Miska
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA.
| |
Collapse
|
13
|
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
|
14
|
Song Y, Wei D, Raza SHA, Zhao Y, Jiang C, Song X, Wu H, Wang X, Luoreng Z, Ma Y. Research progress of intramuscular fat formation based on co-culture. Anim Biotechnol 2023; 34:3216-3236. [PMID: 36200856 DOI: 10.1080/10495398.2022.2127410] [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] [Indexed: 11/01/2022]
Abstract
Intramuscular fat (IMF) is closely related to the meat quality of livestock and poultry. As a new cell culture technique in vitro, cell co-culture has been gradually applied to the related research of IMF formation because it can simulate the changes of microenvironment in vivo during the process of IMF cell formation. In the co-culture model, in addition to studying the effects of skeletal muscle cells on the proliferation and differentiation of IMF, we can also consider the role of many secretion factors in the formation of IMF, thus making the cell research in vitro closer to the real level in vivo. This paper reviewed the generation and origin of IMF, summarized the existing co-culture methods and systems, and discussed the advantages and disadvantages of each method as well as the challenges faced in the establishment of the system, with emphasis on the current status of research on the formation of IMF for human and animal based on co-culture technology.
Collapse
Affiliation(s)
- Yaping Song
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Dawei Wei
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | | | - Yiang Zhao
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Chao Jiang
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Xiaoyu Song
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Hao Wu
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Xingping Wang
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Zhuoma Luoreng
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Yun Ma
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| |
Collapse
|
15
|
Guo B, Shu H, Luo L, Liu X, Ma Y, Zhang J, Liu Z, Zhang Y, Fu L, Song T, Qiao Y, Zhang C. Lactate Conversion by Lactate Dehydrogenase B Is Involved in Beige Adipocyte Differentiation and Thermogenesis in Mice. Nutrients 2023; 15:4846. [PMID: 38004240 PMCID: PMC10674895 DOI: 10.3390/nu15224846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/06/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
Adipose tissue (AT) is the primary reservoir of lipid, the major thermogenesis organ during cold exposure, and an important site for lactate production. However, the utilization of lactate as a metabolic substrate by adipocytes, as well as its potential involvement in the regulation of adipocyte thermogenesis, remain unappreciated. In vitro experiments using primary stromal vascular fraction preadipocytes isolated from mouse inguinal white adipose tissue (iWAT) revealed that lactate dehydrogenase B (LDHB), the key glycolytic enzyme that catalyzes the conversion of lactate to pyruvate, is upregulated during adipocyte differentiation, downregulated upon chronic cold stimulation, and regained after prolonged cold exposure. In addition, the global knockout of Ldhb significantly reduced the masses of iWAT and epididymal WAT (eWAT) and impeded the utilization of iWAT during cold exposure. In addition, Ldhb loss of function impaired the mitochondrial function of iWAT under cold conditions. Together, these findings uncover the involvement of LDHB in adipocyte differentiation and thermogenesis.
Collapse
Affiliation(s)
- Bin Guo
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People’s Hospital), Dongguan 523018, China;
| | - Hui Shu
- Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou 215123, China; (H.S.); (L.L.); (X.L.); (Y.M.); (J.Z.); (Z.L.); (Y.Z.)
| | - Ling Luo
- Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou 215123, China; (H.S.); (L.L.); (X.L.); (Y.M.); (J.Z.); (Z.L.); (Y.Z.)
| | - Xiangpeng Liu
- Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou 215123, China; (H.S.); (L.L.); (X.L.); (Y.M.); (J.Z.); (Z.L.); (Y.Z.)
| | - Yue Ma
- Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou 215123, China; (H.S.); (L.L.); (X.L.); (Y.M.); (J.Z.); (Z.L.); (Y.Z.)
| | - Jie Zhang
- Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou 215123, China; (H.S.); (L.L.); (X.L.); (Y.M.); (J.Z.); (Z.L.); (Y.Z.)
| | - Zhiwei Liu
- Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou 215123, China; (H.S.); (L.L.); (X.L.); (Y.M.); (J.Z.); (Z.L.); (Y.Z.)
| | - Yong Zhang
- Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou 215123, China; (H.S.); (L.L.); (X.L.); (Y.M.); (J.Z.); (Z.L.); (Y.Z.)
| | - Lei Fu
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China;
| | - Tongxing Song
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China;
| | - Yixue Qiao
- Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou 215123, China;
| | - Chi Zhang
- Cambridge-Suda Genomic Resource Center, Soochow University, Suzhou 215123, China; (H.S.); (L.L.); (X.L.); (Y.M.); (J.Z.); (Z.L.); (Y.Z.)
| |
Collapse
|
16
|
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
|
17
|
Nesci S. Proton leak through the UCPs and ANT carriers and beyond: A breath for the electron transport chain. Biochimie 2023; 214:77-85. [PMID: 37336388 DOI: 10.1016/j.biochi.2023.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
Mitochondria produce heat as a result of an ineffective H+ cycling of mitochondria respiration across the inner mitochondrial membrane (IMM). This event present in all mitochondria, known as proton leak, can decrease protonmotive force (Δp) and restore mitochondrial respiration by partially uncoupling the substrate oxidation from the ADP phosphorylation. During impaired conditions of ATP generation with F1FO-ATPase, the Δp increases and IMM is hyperpolarized. In this bioenergetic state, the respiratory complexes support H+ transport until the membrane potential stops the H+ pump activity. Consequently, the electron transfer is stalled and the reduced form of electron carriers of the respiratory chain can generate O2∙¯ triggering the cascade of ROS formation and oxidative stress. The physiological function to attenuate the production of O2∙¯ by Δp dissipation can be attributed to the proton leak supported by the translocases of IMM.
Collapse
Affiliation(s)
- Salvatore Nesci
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, 40064, BO, Italy.
| |
Collapse
|
18
|
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
|
19
|
Zhao JM, Li FQY, Li XY, Jiao DR, Liu XD, Lv XY, Zhao JX. Guanidinoacetic Acid Attenuates Adipogenesis through Regulation of miR-133a in Sheep. Animals (Basel) 2023; 13:3108. [PMID: 37835715 PMCID: PMC10571753 DOI: 10.3390/ani13193108] [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: 08/16/2023] [Revised: 09/30/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
Guanidinoacetic acid (GAA) is an amino acid derivative, previously described in the skeletal muscle of vertebrates, that serves as an important regulator of cellular bioenergetics and has been widely used as a feed additive. Nevertheless, the effect of GAA on adipose tissue growth remains unclear. Here, we hypothesized that dietary GAA negatively affected adipose tissue development in lambs. Lambs were individually fed diets with (0.09%) or without GAA for 70 d ad libitum, and the subcutaneous adipose tissues were sampled for analysis. The results showed that dietary GAA supplementation decreased the girth rib (GR) value (p < 0.01) of lamb carcasses. Both real-time PCR and Western blot analysis suggested that dietary GAA inhibited the expression of adipogenic markers, including peroxisome proliferator-activated receptor γ (PPARγ, p < 0.05), CCAAT/enhancer-binding protein α (C/EBPα, p < 0.01) and sterol-regulatory-element-binding protein 1c (SREBP1C, p < 0.01) in subcutaneous adipose tissue. In vitro, GAA inhibited sheep stromal vascular fraction (SVF) cell proliferation, which was associated with downregulation of proliferating cell nuclear antigen (PCNA, p < 0.05), cyclin-dependent kinase 4 (CDK 4, p < 0.05) and cyclin D1 (p < 0.01). GAA suppressed adipogenesis of SVF cells. Furthermore, miRNA sequencing revealed that GAA affected the miRNA expression profile, and real-time PCR analysis confirmed that miR-133a expression in both subcutaneous adipose tissue and SVF cell was downregulated by GAA. Meanwhile, miR-133a promoted adipogenic differentiation of SVF cells by targeting Sirt1. miR-133a mimics alleviated the inhibitory effect of GAA on SVF cells' adipogenic differentiation. In summary, GAA attenuated adipogenesis of sheep SVF cells, which might occur through miR-133a-modulated Sirt1 expression.
Collapse
Affiliation(s)
- Jia-Min Zhao
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, China (X.-Y.L.); (D.-R.J.)
| | - Fan-Qin-Yu Li
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, China (X.-Y.L.); (D.-R.J.)
| | - Xv-Ying Li
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, China (X.-Y.L.); (D.-R.J.)
| | - Dan-Rong Jiao
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, China (X.-Y.L.); (D.-R.J.)
| | - Xiang-Dong Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Xiao-Yang Lv
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou University, Yangzhou 225009, China
| | - Jun-Xing Zhao
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, China (X.-Y.L.); (D.-R.J.)
| |
Collapse
|
20
|
Ayache L, Bushell A, Lee J, Salminen I, Crespi B. Mother's warmth from maternal genes: genomic imprinting of brown adipose tissue. Evol Med Public Health 2023; 11:379-385. [PMID: 37928960 PMCID: PMC10621903 DOI: 10.1093/emph/eoad031] [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: 05/29/2023] [Revised: 08/04/2023] [Indexed: 11/07/2023] Open
Abstract
Background and objectives Brown adipose tissue (BAT) plays key roles in mammalian physiology, most notably with regard to thermoregulation in infants and juveniles. Previous studies have suggested that intragenomic conflict, in the form of genomic imprinting, mediates BAT thermogenesis, because it represents a public good for groups of siblings, or a mother with her offspring, who huddle together to conserve warmth. By this hypothesis, maternally expressed imprinted genes should promote BAT, while paternally expressed genes should repress it. Methodology We systematically searched the literature using two curated lists of genes imprinted in humans and/or mice, in association with evidence regarding effects of perturbation to imprinted gene expression on BAT development or activity. Results Overall, enhanced BAT was associated with relatively higher expression of maternally expressed imprinted genes, and relatively lower expression of paternally expressed imprinted genes; this pattern was found for 16 of the 19 genes with sufficient information for robust ascertainment (Binomial test, P < 0.005, 2-tailed). Conclusions and implications These results support the kinship theory of imprinting and indicate that future studies of BAT, and its roles in human health and disease, may usefully focus on effects of imprinted genes and associated genomic conflicts.
Collapse
Affiliation(s)
- Lynn Ayache
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Aiden Bushell
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Jessica Lee
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Iiro Salminen
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Bernard Crespi
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| |
Collapse
|
21
|
Heden TD, Franklin MP, Dailey C, Mashek MT, Chen C, Mashek DG. ACOT1 deficiency attenuates high-fat diet-induced fat mass gain by increasing energy expenditure. JCI Insight 2023; 8:e160987. [PMID: 37561578 PMCID: PMC10561717 DOI: 10.1172/jci.insight.160987] [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: 04/13/2022] [Accepted: 08/08/2023] [Indexed: 08/12/2023] Open
Abstract
Acyl-CoA thioesterase 1 (ACOT1) catalyzes the hydrolysis of long-chain acyl-CoAs to free fatty acids and CoA and is typically upregulated in obesity. Whether targeting ACOT1 in the setting of high-fat diet-induced (HFD-induced) obesity would be metabolically beneficial is not known. Here we report that male and female ACOT1KO mice are partially protected from HFD-induced obesity, an effect associated with increased energy expenditure without alterations in physical activity or food intake. In males, ACOT1 deficiency increased mitochondrial uncoupling protein-2 (UCP2) protein abundance while reducing 4-hydroxynonenal, a marker of oxidative stress, in white adipose tissue and liver of HFD-fed mice. Moreover, concurrent knockdown (KD) of UCP2 with ACOT1 in hepatocytes prevented increases in oxygen consumption observed with ACOT1 KD during high lipid loading, suggesting that UCP2-induced uncoupling may increase energy expenditure to attenuate weight gain. Together, these data indicate that targeting ACOT1 may be effective for obesity prevention during caloric excess by increasing energy expenditure.
Collapse
Affiliation(s)
- Timothy D. Heden
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | | | - Christina Dailey
- Department of Biochemistry, Molecular Biology and Biophysics and
| | - Mara T. Mashek
- Department of Biochemistry, Molecular Biology and Biophysics and
| | - Chen Chen
- Department of Biochemistry, Molecular Biology and Biophysics and
| | - Douglas G. Mashek
- Department of Biochemistry, Molecular Biology and Biophysics and
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
22
|
Abstract
In this review, we provide a brief synopsis of the connections between adipose tissue and metabolic health and highlight some recent developments in understanding and exploiting adipocyte biology. Adipose tissue plays critical roles in the regulation of systemic glucose and lipid metabolism and secretes bioactive molecules possessing endocrine, paracrine, and autocrine functions. Dysfunctional adipose tissue has a detrimental impact on metabolic health and is intimately involved in key aspects of metabolic diseases such as insulin resistance, lipid overload, inflammation, and organelle stress. Differences in the distribution of fat depots and adipose characteristics relate to divergent degrees of metabolic dysfunction found in metabolically healthy and unhealthy obese individuals. Thermogenic adipocytes increase energy expenditure via mitochondrial uncoupling or adenosine triphosphate-consuming futile substrate cycles, while functioning as a metabolic sink and participating in crosstalk with other metabolic organs. Manipulation of adipose tissue provides a wealth of opportunities to intervene and combat the progression of associated metabolic diseases. We discuss current treatment modalities for obesity including incretin hormone analogs and touch upon emerging strategies with therapeutic potential including exosome-based therapy, pharmacological activation of brown and beige adipocyte thermogenesis, and administration or inhibition of adipocyte-derived factors.
Collapse
Affiliation(s)
- Sung-Min An
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, CA, USA
| | - Seung-Hee Cho
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, CA, USA
| | - John C. Yoon
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, CA, USA
| |
Collapse
|
23
|
Attardo GM, Benoit JB, Michalkova V, Kondragunta A, Baumann AA, Weiss BL, Malacrida A, Scolari F, Aksoy S. Lipid metabolism dysfunction following symbiont elimination is linked to altered Kennedy pathway homeostasis. iScience 2023; 26:107108. [PMID: 37534171 PMCID: PMC10391724 DOI: 10.1016/j.isci.2023.107108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/27/2023] [Accepted: 06/08/2023] [Indexed: 08/04/2023] Open
Abstract
Lipid metabolism is critical for insect reproduction, especially for species that invest heavily in the early developmental stages of their offspring. The role of symbiotic bacteria during this process is understudied but likely essential. We examined the role of lipid metabolism during the interaction between the viviparous tsetse fly (Glossina morsitans morsitans) and its obligate endosymbiotic bacteria (Wigglesworthia glossinidia) during tsetse pregnancy. We observed increased CTP:phosphocholine cytidylyltransferase (cct1) expression during pregnancy, which is critical for phosphatidylcholine biosynthesis in the Kennedy pathway. Experimental removal of Wigglesworthia impaired lipid metabolism via disruption of the Kennedy pathway, yielding obese mothers whose developing progeny starve. Functional validation via experimental cct1 suppression revealed a phenotype similar to females lacking obligate Wigglesworthia symbionts. These results indicate that, in Glossina, symbiont-derived factors, likely B vitamins, are critical for the proper function of both lipid biosynthesis and lipolysis to maintain tsetse fly fecundity.
Collapse
Affiliation(s)
- Geoffrey M. Attardo
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Department of Entomology and Nematology, Division of Agriculture and Natural Resources, University of California Davis, Davis, CA 95616, USA
| | - Joshua B. Benoit
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Veronika Michalkova
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Section of Molecular and Applied Zoology, Institute of Zoology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Alekhya Kondragunta
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Aaron A. Baumann
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
- Center for Agricultural Synthetic Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Brian L. Weiss
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Anna Malacrida
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
| | - Francesca Scolari
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
- Institute of Molecular Genetics (IGM), Italian National Research Council (CNR), Via Abbiategrasso 207, 27100 Pavia, Italy
| | - Serap Aksoy
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| |
Collapse
|
24
|
Jin L, Han S, Lv X, Li X, Zhang Z, Kuang H, Chen Z, Lv CA, Peng W, Yang Z, Yang M, Mi L, Liu T, Ma S, Qiu X, Wang Q, Pan X, Shan P, Feng Y, Li J, Wang F, Xie L, Zhao X, Fu JF, Lin JD, Meng ZX. The muscle-enriched myokine Musclin impairs beige fat thermogenesis and systemic energy homeostasis via Tfr1/PKA signaling in male mice. Nat Commun 2023; 14:4257. [PMID: 37468484 PMCID: PMC10356794 DOI: 10.1038/s41467-023-39710-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/27/2023] [Indexed: 07/21/2023] Open
Abstract
Skeletal muscle and thermogenic adipose tissue are both critical for the maintenance of body temperature in mammals. However, whether these two tissues are interconnected to modulate thermogenesis and metabolic homeostasis in response to thermal stress remains inconclusive. Here, we report that human and mouse obesity is associated with elevated Musclin levels in both muscle and circulation. Intriguingly, muscle expression of Musclin is markedly increased or decreased when the male mice are housed in thermoneutral or chronic cool conditions, respectively. Beige fat is then identified as the primary site of Musclin action. Muscle-transgenic or AAV-mediated overexpression of Musclin attenuates beige fat thermogenesis, thereby exacerbating diet-induced obesity and metabolic disorders in male mice. Conversely, Musclin inactivation by muscle-specific ablation or neutralizing antibody treatment promotes beige fat thermogenesis and improves metabolic homeostasis in male mice. Mechanistically, Musclin binds to transferrin receptor 1 (Tfr1) and antagonizes Tfr1-mediated cAMP/PKA-dependent thermogenic induction in beige adipocytes. This work defines the temperature-sensitive myokine Musclin as a negative regulator of adipose thermogenesis that exacerbates the deterioration of metabolic health in obese male mice and thus provides a framework for the therapeutic targeting of this endocrine pathway.
Collapse
Affiliation(s)
- Lu Jin
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuang Han
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
- Department of Geriatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Chronic Disease Research Institute, Zhejiang University School of Public Health, Hangzhou, China
| | - Xue Lv
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
- Chronic Disease Research Institute, Zhejiang University School of Public Health, Hangzhou, China
| | - Xiaofei Li
- Department of Sport Medicine, The Lianyungang First People's Hospital, Affiliated Hospital of Xuzhou Medical University, Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, China
| | - Ziyin Zhang
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
- Chronic Disease Research Institute, Zhejiang University School of Public Health, Hangzhou, China
| | - Henry Kuang
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, USA
| | - Zhimin Chen
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, USA
| | - Cheng-An Lv
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Peng
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhuoying Yang
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
- Chronic Disease Research Institute, Zhejiang University School of Public Health, Hangzhou, China
| | - Miqi Yang
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
- Chronic Disease Research Institute, Zhejiang University School of Public Health, Hangzhou, China
| | - Lin Mi
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, USA
| | - Tongyu Liu
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, USA
| | - Shengshan Ma
- Department of Sport Medicine, The Lianyungang First People's Hospital, Affiliated Hospital of Xuzhou Medical University, Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, China
| | - Xinyuan Qiu
- Department of Biology and Chemistry, College of Science, National University of Defense Technology, Changsha, China
| | - Qintao Wang
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
- Chronic Disease Research Institute, Zhejiang University School of Public Health, Hangzhou, China
| | - Xiaowen Pan
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pengfei Shan
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Feng
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jin Li
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Liwei Xie
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Xuyun Zhao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun-Fen Fu
- Department of Endocrinology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jiandie D Lin
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, USA
| | - Zhuo-Xian Meng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Geriatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Chronic Disease Research Institute, Zhejiang University School of Public Health, Hangzhou, China.
| |
Collapse
|
25
|
Cal K, Leyva A, Rodríguez-Duarte J, Ruiz S, Santos L, Colella L, Ingold M, Vilaseca C, Galliussi G, Ziegler L, Peclat TR, Bresque M, Handy RM, King R, dos Reis LM, Espasandin C, Breining P, Dapueto R, Lopez A, Thompson KL, Agorrody G, DeVallance E, Meadows E, Lewis SE, Barbosa GCS, de Souza LOL, Chichierchio MS, Valez V, Aicardo A, Contreras P, Vendelbo MH, Jakobsen S, Kamaid A, Porcal W, Calliari A, Verdes JM, Du J, Wang Y, Hollander JM, White TA, Radi R, Moyna G, Quijano C, O’Doherty R, Moraes-Vieira P, Holloway GP, Leonardi R, Mori MA, Camacho-Pereira J, Kelley EE, Duran R, Lopez GV, Batthyány C, Chini EN, Escande C. A nitroalkene derivative of salicylate alleviates diet-induced obesity by activating creatine metabolism and non-shivering thermogenesis. RESEARCH SQUARE 2023:rs.3.rs-3101395. [PMID: 37502859 PMCID: PMC10371099 DOI: 10.21203/rs.3.rs-3101395/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Obesity-related type II diabetes (diabesity) has increased global morbidity and mortality dramatically. Previously, the ancient drug salicylate demonstrated promise for the treatment of type II diabetes, but its clinical use was precluded due to high dose requirements. In this study, we present a nitroalkene derivative of salicylate, 5-(2-nitroethenyl)salicylic acid (SANA), a molecule with unprecedented beneficial effects in diet-induced obesity (DIO). SANA reduces DIO, liver steatosis and insulin resistance at doses up to 40 times lower than salicylate. Mechanistically, SANA stimulated mitochondrial respiration and increased creatine-dependent energy expenditure in adipose tissue. Indeed, depletion of creatine resulted in the loss of SANA action. Moreover, we found that SANA binds to creatine kinases CKMT1/2, and downregulation CKMT1 interferes with the effect of SANA in vivo. Together, these data demonstrate that SANA is a first-in-class activator of creatine-dependent energy expenditure and thermogenesis in adipose tissue and emerges as a candidate for the treatment of diabesity.
Collapse
Affiliation(s)
- Karina Cal
- Laboratory of Metabolic Diseases and Aging, Institut Pasteur Montevideo, Uruguay
- Unidad Biofísica, Departamento de Biociencias, Facultad de Veterinaria, Udelar, Uruguay
| | - Alejandro Leyva
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur Montevideo, Uruguay
- Unidad de Bioquímica y Proteómica Analíticas, Institut Pasteur de Montevideo, IIBCE, Uruguay
| | - Jorge Rodríguez-Duarte
- Laboratory of Metabolic Diseases and Aging, Institut Pasteur Montevideo, Uruguay
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur Montevideo, Uruguay
| | - Santiago Ruiz
- Laboratory of Metabolic Diseases and Aging, Institut Pasteur Montevideo, Uruguay
| | - Leonardo Santos
- Laboratory of Metabolic Diseases and Aging, Institut Pasteur Montevideo, Uruguay
| | - Lucía Colella
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur Montevideo, Uruguay
- Departamento de Química Orgánica, Facultad de Química, Udelar, Uruguay
| | - Mariana Ingold
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur Montevideo, Uruguay
- Departamento de Química Orgánica, Facultad de Química, Udelar, Uruguay
| | - Cecilia Vilaseca
- Departamento de Fisiología, Facultad de Medicina, Udelar, Uruguay
| | - German Galliussi
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur Montevideo, Uruguay
- Laboratory of Immunoregulation and Inflammation; Institut Pasteur Montevideo, Uruguay
| | - Lucía Ziegler
- Departamento de Ecología y Gestión Ambiental, Centro Universitario Regional del Este, Udelar, Maldonado, Uruguay
| | - Thais R. Peclat
- Mayo Clinic Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering; Mayo Clinic, Rochester, MN, USA
| | - Mariana Bresque
- Laboratory of Metabolic Diseases and Aging, Institut Pasteur Montevideo, Uruguay
| | - Rachel M Handy
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Rachel King
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown WV, USA
| | - Larissa Menezes dos Reis
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil; Obesity and Comorbidities Research Center (OCRC), University of Campinas, SP, Brazil; Experimental Medicine Research Cluster (EMRC), University of Campinas, SP, Brazil
| | - Camila Espasandin
- Laboratory of Metabolic Diseases and Aging, Institut Pasteur Montevideo, Uruguay
- Unidad Bioquìmica, Facultad de Veterinaria, Udelar, Uruguay
| | | | - Rosina Dapueto
- Laboratory of Metabolic Diseases and Aging, Institut Pasteur Montevideo, Uruguay
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur Montevideo, Uruguay
- Área I+D Biomédico, CUDIM, Uruguay
| | - Andrés Lopez
- Laboratorio de Fisicoquímica Orgánica, Departamento de Química del Litoral, CENUR Litoral Norte, Udelar, Uruguay
| | - Katie L. Thompson
- Mayo Clinic Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering; Mayo Clinic, Rochester, MN, USA
| | - Guillermo Agorrody
- Departamento de Fisiopatología, Hospital de Clínicas, Facultad de Medicina, Udelar, Uruguay
| | - Evan DeVallance
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Ethan Meadows
- Mitochondria, Metabolism and Bioenergetics Working Group; School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Sara E. Lewis
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, Morgantown, WV, USA
- Mitochondria, Metabolism and Bioenergetics Working Group; School of Medicine, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, USA
| | - Gabriele Catarine Santana Barbosa
- Laboratory of Bioenergetics and Mitochondrial Physiology, Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Brazil
| | - Leonardo Osbourne Lai de Souza
- Laboratory of Bioenergetics and Mitochondrial Physiology, Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Brazil
| | - Marina Santos Chichierchio
- Laboratory of Bioenergetics and Mitochondrial Physiology, Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Brazil
| | - Valeria Valez
- Cátedra de Bioquímica y Biofísica, Facultad de Odontología, Udelar, Uruguay
- Centro de Investigaciones Biomédicas (CEINBIO), Udelar, Uruguay
- Departamento de Bioquímica, Facultad de Medicina, Udelar, Uruguay
| | - Adrián Aicardo
- Centro de Investigaciones Biomédicas (CEINBIO), Udelar, Uruguay
- Departamento de Bioquímica, Facultad de Medicina, Udelar, Uruguay
- Departamento de Nutrición Clínica, Escuela de Nutrición, Udelar, Uruguay
| | - Paola Contreras
- Laboratory of Metabolic Diseases and Aging, Institut Pasteur Montevideo, Uruguay
- Departamento de Fisiología, Facultad de Medicina, Udelar, Uruguay
| | - Mikkel H. Vendelbo
- Department of Biomedicine, Aarhus University, Denmark
- Department of Nuclear Medicine and PET, Aarhus University Hospital, Denmark
| | - Steen Jakobsen
- Department of Nuclear Medicine and PET, Aarhus University Hospital, Denmark
| | - Andrés Kamaid
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur Montevideo, Uruguay
- Unidad de Bioquímica y Proteómica Analíticas, Institut Pasteur de Montevideo, IIBCE, Uruguay
- Unidad de Bioimagenología Avanzada. Institut Pasteur de Montevideo, Uruguay
| | - Williams Porcal
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur Montevideo, Uruguay
- Departamento de Química Orgánica, Facultad de Química, Udelar, Uruguay
| | - Aldo Calliari
- Laboratory of Metabolic Diseases and Aging, Institut Pasteur Montevideo, Uruguay
- Unidad Biofísica, Departamento de Biociencias, Facultad de Veterinaria, Udelar, Uruguay
| | - José Manuel Verdes
- Unidad Patología, Departamento de Patobiología; Facultad de Veterinaria, Udelar, Uruguay
| | - Jianhai Du
- Mitochondria, Metabolism and Bioenergetics Working Group; School of Medicine, West Virginia University, Morgantown, WV, USA
- Department of Ophthalmology and Visual Sciences, Department of Biochemistry, West Virginia University, Morgantown, USA
| | - Yekai Wang
- Department of Ophthalmology and Visual Sciences, Department of Biochemistry, West Virginia University, Morgantown, USA
| | - John M Hollander
- Mitochondria, Metabolism and Bioenergetics Working Group; School of Medicine, West Virginia University, Morgantown, WV, USA
- Division of Exercise Physiology, West Virginia University, Morgantown, USA
| | - Thomas A. White
- Mayo Clinic Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Rafael Radi
- Centro de Investigaciones Biomédicas (CEINBIO), Udelar, Uruguay
- Departamento de Bioquímica, Facultad de Medicina, Udelar, Uruguay
| | - Guillermo Moyna
- Laboratorio de Fisicoquímica Orgánica, Departamento de Química del Litoral, CENUR Litoral Norte, Udelar, Uruguay
| | - Celia Quijano
- Centro de Investigaciones Biomédicas (CEINBIO), Udelar, Uruguay
- Departamento de Bioquímica, Facultad de Medicina, Udelar, Uruguay
| | - Robert O’Doherty
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pennsylvania
- Department of Microbiology and Molecular Genetics; University of Pittsburgh, Pennsylvania
| | - Pedro Moraes-Vieira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, SP, Brazil; Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil; Obesity and Comorbidities Research Center (OCRC), University of Campinas, SP, Brazil; Experimental Medicine Research Cluster (EMRC), University of Campinas, SP, Brazil
| | - Graham P Holloway
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Roberta Leonardi
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown WV, USA
- Mitochondria, Metabolism and Bioenergetics Working Group; School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Marcelo A Mori
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, SP, Brazil; Obesity and Comorbidities Research Center (OCRC), Campinas, SP, Brazil; Experimental Medicine Research Cluster (EMRC), Campinas, SP, Brazil; Instituto Nacional de Obesidade e Diabetes, Campinas, SP, Brazil
| | - Juliana Camacho-Pereira
- Laboratory of Bioenergetics and Mitochondrial Physiology, Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Brazil
| | - Eric E. Kelley
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, Morgantown, WV, USA
- Mitochondria, Metabolism and Bioenergetics Working Group; School of Medicine, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, USA
| | - Rosario Duran
- Unidad de Bioquímica y Proteómica Analíticas, Institut Pasteur de Montevideo, IIBCE, Uruguay
| | - Gloria V. Lopez
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur Montevideo, Uruguay
- Departamento de Química Orgánica, Facultad de Química, Udelar, Uruguay
| | - Carlos Batthyány
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur Montevideo, Uruguay
| | - Eduardo N. Chini
- Mayo Clinic Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering; Mayo Clinic, Rochester, MN, USA
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Carlos Escande
- Laboratory of Metabolic Diseases and Aging, Institut Pasteur Montevideo, Uruguay
| |
Collapse
|
26
|
Fu T, Sun W, Xue J, Zhou Z, Wang W, Guo Q, Chen X, Zhou D, Xu Z, Liu L, Xiao L, Mao Y, Yang L, Yin Y, Zhang XN, Wan Q, Lu B, Chen Y, Zhu MS, Scherer PE, Fang L, Piao HL, Shao M, Gan Z. Proteolytic rewiring of mitochondria by LONP1 directs cell identity switching of adipocytes. Nat Cell Biol 2023; 25:848-864. [PMID: 37217599 DOI: 10.1038/s41556-023-01155-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 04/19/2023] [Indexed: 05/24/2023]
Abstract
Mitochondrial proteases are emerging as key regulators of mitochondrial plasticity and acting as both protein quality surveillance and regulatory enzymes by performing highly regulated proteolytic reactions. However, it remains unclear whether the regulated mitochondrial proteolysis is mechanistically linked to cell identity switching. Here we report that cold-responsive mitochondrial proteolysis is a prerequisite for white-to-beige adipocyte cell fate programming during adipocyte thermogenic remodelling. Thermogenic stimulation selectively promotes mitochondrial proteostasis in mature white adipocytes via the mitochondrial protease LONP1. Disruption of LONP1-dependent proteolysis substantially impairs cold- or β3 adrenergic agonist-induced white-to-beige identity switching of mature adipocytes. Mechanistically, LONP1 selectively degrades succinate dehydrogenase complex iron sulfur subunit B and ensures adequate intracellular succinate levels. This alters the histone methylation status on thermogenic genes and thereby enables adipocyte cell fate programming. Finally, augmented LONP1 expression raises succinate levels and corrects ageing-related impairments in white-to-beige adipocyte conversion and adipocyte thermogenic capacity. Together, these findings reveal that LONP1 links proteolytic surveillance to mitochondrial metabolic rewiring and directs cell identity conversion during adipocyte thermogenic remodelling.
Collapse
Affiliation(s)
- Tingting Fu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Wanping Sun
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Jiachen Xue
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Zheng Zhou
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Wen Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Qiqi Guo
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Xinyi Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Danxia Zhou
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Zhisheng Xu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Lin Liu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Liwei Xiao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Yan Mao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Likun Yang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Yujing Yin
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Xue-Na Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Qiangyou Wan
- CAS Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Chinese Academy of Sciences, Shanghai, China
| | - Bin Lu
- Hengyang Medical College, University of South China, Hengyang, China
| | - Yuncong Chen
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
| | - Min-Sheng Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Philipp E Scherer
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lei Fang
- Jiangsu Key Laboratory of Molecular Medicine, Chemistry and Biomedicine Innovation Center (ChemBIC), Medical School of Nanjing University, Nanjing, China
| | - Hai-Long Piao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Mengle Shao
- CAS Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Chinese Academy of Sciences, Shanghai, China.
| | - Zhenji Gan
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China.
- Jiangsu Key Laboratory of Molecular Medicine, Chemistry and Biomedicine Innovation Center (ChemBIC), Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
27
|
Liu Y, Sun K, Gan Y, Liu H, Yu J, Xu W, Zhang L, Chen D. RNA-Sequencing Reveals Gene Expression and Pathway Signatures in Umbilical Cord Blood Affected by Birth Delivery Mode. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:228-242. [PMID: 37325709 PMCID: PMC10260732 DOI: 10.1007/s43657-022-00086-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/11/2022] [Accepted: 11/16/2022] [Indexed: 06/17/2023]
Abstract
Cesarean section (CS) confers increased risk of type I diabetes, asthma, inflammatory bowel disease, celiac disease, overweight and obesity, etc., in the offspring. However, the underlying mechanism remains unknown. To investigate the influence of CS on gene expression in cord blood, we have performed RNA-sequencing followed by single-gene analysis, gene set enrichment analysis, gene co-expression network analysis, and interacting genes/proteins analysis in eight full-term infants born by elective CS and eight matched vaginally delivered (VD) infants. Crucial genes identified above were further validated in another 20 CS and 20 VD infants. We found for the first time that mRNA expression of genes involved in immune response (IL12A, INFG, IL1B, TNF, MIF, IL4, CA1, IFI27, HLA-DOB and EPHB1) and metabolism (DLK1, CYP2A6 and GATM) were significantly influenced by CS. Notably, serum TNF-α and IFN-γ were remarkably up-regulated in the CS infants (p = 5.0 × 10-4 and 3.0 × 10-3, respectively) compared to the VD infants. It is biologically plausible that CS may exert adverse impacts on offspring health through influencing expression of genes in the above processes. These findings will help understand the potential underlying mechanisms of the adverse health impacts of CS and identify biomarkers for future health of offspring born with different delivery modes. Supplementary Information The online version contains supplementary material available at 10.1007/s43657-022-00086-7.
Collapse
Affiliation(s)
- Yongjie Liu
- Ministry of Education and Shanghai Key Laboratory of Children’s Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092 China
| | - Kun Sun
- Shenzhen Bay Laboratory, Shenzhen, 518107 China
| | - Yuexin Gan
- Ministry of Education and Shanghai Key Laboratory of Children’s Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092 China
| | - Han Liu
- Department of Obstetrics and Gynaecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025 China
| | - Juehua Yu
- Ministry of Education and Shanghai Key Laboratory of Children’s Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092 China
- Centre for Experimental Studies and Research, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032 China
| | - Wei Xu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032 China
| | - Lin Zhang
- Ministry of Education and Shanghai Key Laboratory of Children’s Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092 China
- Department of Obstetrics and Gynaecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025 China
| | - Dan Chen
- Department of Gynecology, Third Xiangya Hospital of Central South University, Changsha, 410013 China
- Shanghai Fosun Pharmaceutical Industrial Development, Co., Ltd, Shanghai, 200233 China
| |
Collapse
|
28
|
Abstract
The circadian clock plays an essential role in coordinating feeding and metabolic rhythms with the light/dark cycle. Disruption of clocks is associated with increased adiposity and metabolic disorders, whereas aligning feeding time with cell-autonomous rhythms in metabolism improves health. Here, we provide a comprehensive overview of recent literature in adipose tissue biology as well as our understanding of molecular mechanisms underlying the circadian regulation of transcription, metabolism, and inflammation in adipose tissue. We highlight recent efforts to uncover the mechanistic links between clocks and adipocyte metabolism, as well as its application to dietary and behavioral interventions to improve health and mitigate obesity.
Collapse
Affiliation(s)
- Chelsea Hepler
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| |
Collapse
|
29
|
Huang J, Rao L, Zhang W, Chen X, Li H, Zhang F, Xie J, Wei Q. Effect of crossbreeding and sex on slaughter performance and meat quality in Xingguo gray goose based on multiomics data analysis. Poult Sci 2023; 102:102753. [PMID: 37267641 PMCID: PMC10244692 DOI: 10.1016/j.psj.2023.102753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/17/2023] [Accepted: 04/23/2023] [Indexed: 06/04/2023] Open
Abstract
Here, we examined the effects of crossbreeding and sex on growth performance, slaughter performance, and meat quality in Xingguo gray (XG) goose, using transcriptomic and metabolomic techniques. The experiment was conducted using 400 goslings (1-day old) of 2 genotypes: the XG breed and its ternary hybrids [F2 geese; (XG Goose♂ × Yangzhou Goose♀)♀ × Shitou Goose♂]. The goslings were divided into 4 groups: female XG, male XG, female F2 geese, and male F2 geese, and growth parameters were examined at 70 d of age, using 30 birds from each group. Following slaughter, samples of breast and thigh muscles were collected from each group for chemical, metabolome, and transcriptome analyses. Growth rate, live body and slaughter weights, meat chemical composition, and muscle fiber diameter were affected by crossbreeding and sex. Crossbreeding significantly improved the dressing percentage, semieviscerated rate, eviscerated yield, and abdominal fat yield of XG geese. To clarify the potential regulatory network affected by crossbreeding and sex, we used RNA-seq and nontargeted metabolomics to detect changes in male and female goose breast muscle. The transcriptome results showed that there were 534, 323, 297, and 492 differently expressed genes (DEGs) among the 4 comparison groups (XG-Female vs. F2-Female, XG-Male vs. F2-Male, F2-Male vs. F2-Female, and XG-Male vs. XG-Female, respectively) that were mainly related to muscle growth and development and fatty acid metabolism pathways. A total of 141 significantly differentially accumulated metabolites (DAMs) were enriched in serine and threonine, propionate, and pyruvate metabolism. Finally, we comprehensively analyzed the metabolome and transcriptome data and found that many DEGs and DAMs played crucial roles in lipid metabolism and muscle growth and development. In summary, crossbreeding can improve XG goose production performance and affect breast muscle gene expression and metabolites in both female and male geese.
Collapse
Affiliation(s)
- Jiangnan Huang
- Institute of Animal Husbandry and Veterinary Medicine, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, China
| | - Linjie Rao
- Institute of Animal Husbandry and Veterinary Medicine, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, China
| | - Weihong Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, China
| | - Xiaolian Chen
- Institute of Animal Husbandry and Veterinary Medicine, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, China
| | - Haiqin Li
- Institute of Animal Husbandry and Veterinary Medicine, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, China
| | - Fanfan Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, China
| | - Jinfang Xie
- Institute of Animal Husbandry and Veterinary Medicine, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, China
| | - Qipeng Wei
- Institute of Animal Husbandry and Veterinary Medicine, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, China.
| |
Collapse
|
30
|
Coulter AA, Greenway FL, Zhang D, Ghosh S, Coulter CR, James SL, He Y, Cusimano LA, Rebello CJ. Naringenin and β-carotene convert human white adipocytes to a beige phenotype and elevate hormone- stimulated lipolysis. Front Endocrinol (Lausanne) 2023; 14:1148954. [PMID: 37143734 PMCID: PMC10153092 DOI: 10.3389/fendo.2023.1148954] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/20/2023] [Indexed: 05/06/2023] Open
Abstract
Introduction Naringenin, a peroxisome proliferator-activated receptor (PPAR) activator found in citrus fruits, upregulates markers of thermogenesis and insulin sensitivity in human adipose tissue. Our pharmacokinetics clinical trial demonstrated that naringenin is safe and bioavailable, and our case report showed that naringenin causes weight loss and improves insulin sensitivity. PPARs form heterodimers with retinoic-X-receptors (RXRs) at promoter elements of target genes. Retinoic acid is an RXR ligand metabolized from dietary carotenoids. The carotenoid β-carotene reduces adiposity and insulin resistance in clinical trials. Our goal was to examine if carotenoids strengthen the beneficial effects of naringenin on human adipocyte metabolism. Methods Human preadipocytes from donors with obesity were differentiated in culture and treated with 8µM naringenin + 2µM β-carotene (NRBC) for seven days. Candidate genes involved in thermogenesis and glucose metabolism were measured as well as hormone-stimulated lipolysis. Results We found that β-carotene acts synergistically with naringenin to boost UCP1 and glucose metabolism genes including GLUT4 and adiponectin, compared to naringenin alone. Protein levels of PPARα, PPARγ and PPARγ-coactivator-1α, key modulators of thermogenesis and insulin sensitivity, were also upregulated after treatment with NRBC. Transcriptome sequencing was conducted and the bioinformatics analyses of the data revealed that NRBC induced enzymes for several non-UCP1 pathways for energy expenditure including triglyceride cycling, creatine kinases, and Peptidase M20 Domain Containing 1 (PM20D1). A comprehensive analysis of changes in receptor expression showed that NRBC upregulated eight receptors that have been linked to lipolysis or thermogenesis including the β1-adrenergic receptor and the parathyroid hormone receptor. NRBC increased levels of triglyceride lipases and agonist-stimulated lipolysis in adipocytes. We observed that expression of RXRγ, an isoform of unknown function, was induced ten-fold after treatment with NRBC. We show that RXRγ is a coactivator bound to the immunoprecipitated PPARγ protein complex from white and beige human adipocytes. Discussion There is a need for obesity treatments that can be administered long-term without side effects. NRBC increases the abundance and lipolytic response of multiple receptors for hormones released after exercise and cold exposure. Lipolysis provides the fuel for thermogenesis, and these observations suggest that NRBC has therapeutic potential.
Collapse
Affiliation(s)
- Ann A. Coulter
- Computational Biology, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Frank L. Greenway
- Clinical Trials, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Dachuan Zhang
- Biostatistics, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Sujoy Ghosh
- Adjunct Faculty, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Cathryn R. Coulter
- Computational Biology, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Sarah L. James
- Computational Biology, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Yanlin He
- Brain Glycemic and Metabolism Control, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Luke A. Cusimano
- Cusimano Plastic and Reconstructive Surgery, Baton Rouge, LA, United States
| | - Candida J. Rebello
- Nutrition and Chronic Disease, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| |
Collapse
|
31
|
Haneishi Y, Furuya Y, Hasegawa M, Takemae H, Tanioka Y, Mizutani T, Rossi M, Miyamoto J. Polyunsaturated fatty acids-rich dietary lipid prevents high fat diet-induced obesity in mice. Sci Rep 2023; 13:5556. [PMID: 37019935 PMCID: PMC10076282 DOI: 10.1038/s41598-023-32851-7] [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: 11/16/2022] [Accepted: 04/03/2023] [Indexed: 04/07/2023] Open
Abstract
Diet is the primary factor affecting host nutrition and metabolism, with excess food intake, especially high-calorie diets, such as high-fat and high-sugar diets, causing an increased risk of obesity and related disorders. Obesity alters the gut microbial composition and reduces microbial diversity and causes changes in specific bacterial taxa. Dietary lipids can alter the gut microbial composition in obese mice. However, the regulation of gut microbiota and host energy homeostasis by different polyunsaturated fatty acids (PUFAs) in dietary lipids remains unknown. Here, we demonstrated that different PUFAs in dietary lipids improved host metabolism in high-fat diet (HFD)-induced obesity in mice. The intake of the different PUFA-enriched dietary lipids improved metabolism in HFD-induced obesity by regulating glucose tolerance and inhibiting colonic inflammation. Moreover, the gut microbial compositions were different among HFD and modified PUFA-enriched HFD-fed mice. Thus, we have identified a new mechanism underlying the function of different PUFAs in dietary lipids in regulating host energy homeostasis in obese conditions. Our findings shed light on the prevention and treatment of metabolic disorders by targeting the gut microbiota.
Collapse
Affiliation(s)
- Yuri Haneishi
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, 183-8509, Japan
| | - Yuma Furuya
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, 183-8509, Japan
| | - Mayu Hasegawa
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, 183-8509, Japan
| | - Hitoshi Takemae
- Center for Infectious Diseases Epidemiology and Prevention Research: CEPiR, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, 183-8509, Japan
| | - Yuri Tanioka
- Department of International Food and Agricultural Science, Faculty of International Food and Agricultural Studies, Tokyo University of Agriculture, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Tetsuya Mizutani
- Center for Infectious Diseases Epidemiology and Prevention Research: CEPiR, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, 183-8509, Japan
| | - Mauro Rossi
- Institute of Food Sciences, CNR, via Roma 64, 83100, Avellino, Italy
| | - Junki Miyamoto
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, 183-8509, Japan.
| |
Collapse
|
32
|
Zhu L, Zhang J, Yang H, Li G, Li H, Deng Z, Zhang B. Propolis polyphenols: A review on the composition and anti-obesity mechanism of different types of propolis polyphenols. Front Nutr 2023; 10:1066789. [PMID: 37063322 PMCID: PMC10102383 DOI: 10.3389/fnut.2023.1066789] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 03/14/2023] [Indexed: 04/03/2023] Open
Abstract
Obesity, one of the most common nutritional diseases worldwide, can lead to dyslipidemia, high blood sugar, high blood pressure, and inflammation. Some drugs have been developed to ameliorate obesity. However, these drugs may cause serious side effects. Therefore, there is an urgent need for alternative “natural” remedies including propolis. Studies have found that propolis has excellent anti-obesity activity in in vitro and in vivo models during the past decades, of which polyphenols are the key component in regulating weight loss. This review focused on the different polyphenol compositions of propolis from different regions and plants, the evidence for the anti-obesity effects of different types of propolis and its derivatives, discussed the impact of propolis polyphenols on obesity related signal pathways, and proposed the molecular mechanism of how propolis polyphenols affect these signal pathways. For example, propolis and its derivatives regulate lipid metabolism related proteins, such as PPARα, PPARγ, SREBP-1&2, and HMG CoA etc., destroy the formation of CREB/CRTC2 transcription complex, activate Nrf2 pathway or inhibit protein kinase IKK ε/TBK1, thereby affecting fat production and lipid metabolism; The effects of propolis on adipokines (adiponectin, leptin and inflammatory factors) were discussed. Additionally, the mechanism of polyphenols in propolis promoting the browning of adipose tissues and the relationship between intestinal microorganisms was summarized. These information may be of value to better understand how specific propolis polyphenols interact with specific signaling pathways and help guide the development of new drugs to combat obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Liuying Zhu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Jinwu Zhang
- Nanchang Concentric Purple Nest Biological Engineering Co., Ltd., Nanchang, China
| | - Hui Yang
- Nanchang Concentric Purple Nest Biological Engineering Co., Ltd., Nanchang, China
| | - Guangyan Li
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
- Nanchang Concentric Purple Nest Biological Engineering Co., Ltd., Nanchang, China
| | - Hongyan Li
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Bing Zhang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
- *Correspondence: Bing Zhang,
| |
Collapse
|
33
|
Villanueva-Carmona T, Cedó L, Madeira A, Ceperuelo-Mallafré V, Rodríguez-Peña MM, Núñez-Roa C, Maymó-Masip E, Repollés-de-Dalmau M, Badia J, Keiran N, Mirasierra M, Pimenta-Lopes C, Sabadell-Basallote J, Bosch R, Caubet L, Escolà-Gil JC, Fernández-Real JM, Vilarrasa N, Ventura F, Vallejo M, Vendrell J, Fernández-Veledo S. SUCNR1 signaling in adipocytes controls energy metabolism by modulating circadian clock and leptin expression. Cell Metab 2023; 35:601-619.e10. [PMID: 36977414 DOI: 10.1016/j.cmet.2023.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/21/2022] [Accepted: 03/03/2023] [Indexed: 03/30/2023]
Abstract
Adipose tissue modulates energy homeostasis by secreting leptin, but little is known about the factors governing leptin production. We show that succinate, long perceived as a mediator of immune response and lipolysis, controls leptin expression via its receptor SUCNR1. Adipocyte-specific deletion of Sucnr1 influences metabolic health according to nutritional status. Adipocyte Sucnr1 deficiency impairs leptin response to feeding, whereas oral succinate mimics nutrient-related leptin dynamics via SUCNR1. SUCNR1 activation controls leptin expression via the circadian clock in an AMPK/JNK-C/EBPα-dependent manner. Although the anti-lipolytic role of SUCNR1 prevails in obesity, its function as a regulator of leptin signaling contributes to the metabolically favorable phenotype in adipocyte-specific Sucnr1 knockout mice under standard dietary conditions. Obesity-associated hyperleptinemia in humans is linked to SUCNR1 overexpression in adipocytes, which emerges as the major predictor of adipose tissue leptin expression. Our study establishes the succinate/SUCNR1 axis as a metabolite-sensing pathway mediating nutrient-related leptin dynamics to control whole-body homeostasis.
Collapse
Affiliation(s)
- Teresa Villanueva-Carmona
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Lídia Cedó
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Ana Madeira
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Victòria Ceperuelo-Mallafré
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain; Department of Medicine and Surgery, Universitat Rovira i Virgili (URV), Reus 43201, Spain
| | - M-Mar Rodríguez-Peña
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Catalina Núñez-Roa
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Elsa Maymó-Masip
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Maria Repollés-de-Dalmau
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain; Department of Medicine and Surgery, Universitat Rovira i Virgili (URV), Reus 43201, Spain
| | - Joan Badia
- Institut d'Oncologia de la Catalunya Sud, Hospital Universitari Sant Joan de Reus, IISPV, Reus 43204, Spain
| | - Noelia Keiran
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Mercedes Mirasierra
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain; Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid (CSIC/UAM), Madrid 28029, Spain
| | - Carolina Pimenta-Lopes
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Joan Sabadell-Basallote
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Ramón Bosch
- Department of Pathology, Oncological Pathology and Bioinformatics Research Group, Hospital de Tortosa Verge de la Cinta, IISPV, Tortosa 43500, Spain
| | - Laura Caubet
- General and Digestive Surgery Service, Hospital Sant Pau i Santa Tecla, Institut d'Investigació Sanitària Pere Virgili, Tarragona 43003, Spain
| | - Joan Carles Escolà-Gil
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain; Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona 08041, Spain; Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona 08193, Spain
| | - José-Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), Salt 17190, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CB06/03/010), Instituto de Salud Carlos III, Madrid 28029, Spain; Department of Medical Sciences, School of Medicine, University of Girona, Girona 17004, Spain
| | - Nuria Vilarrasa
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain; Department of Endocrinology and Nutrition, Hospital Universitari Bellvitge - IDIBELL, Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Francesc Ventura
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Mario Vallejo
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain; Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid (CSIC/UAM), Madrid 28029, Spain
| | - Joan Vendrell
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain; Department of Medicine and Surgery, Universitat Rovira i Virgili (URV), Reus 43201, Spain
| | - Sonia Fernández-Veledo
- Department of Endocrinology and Nutrition, Research Unit, Institut d'Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari de Tarragona Joan XXIII, Tarragona 43005, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid 28029, Spain.
| |
Collapse
|
34
|
Choi M, Yun JW. β-Carotene induces UCP1-independent thermogenesis via ATP-consuming futile cycles in 3T3-L1 white adipocytes. Arch Biochem Biophys 2023; 739:109581. [PMID: 36948352 DOI: 10.1016/j.abb.2023.109581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/17/2023] [Accepted: 03/19/2023] [Indexed: 03/24/2023]
Abstract
The activation of brown fat and induction of beige adipocytes, so-called non-shivering thermogenesis, is emerging as a promising target for therapeutic intervention in obesity management. Our previous report demonstrated that β-carotene (BC) induces beige adipocytes to increase UCP1-dependent thermogenic activity. However, the UCP1-independent thermogenic effect of BC on adipose tissues remains unexplored. In this study, we examined the effects of BC on UCP1-independent thermogenic activity with a focus on the ATP-consuming futile cycles in 3T3-L1 adipocytes. BC increased intracellular calcium levels and stimulated the expression of calcium cycling-related proteins, including sarcoendoplasmic reticulum Ca2+-ATPase (SERCA) 2b, ryanodine receptor 2 (RyR2), voltage-dependent anion channel (VDAC), mitochondrial calcium uniporter (MCU), and Ca2+/calmodulin-dependent protein kinase 2 (CaMK2) in 3T3-L1 white adipocytes. In addition, BC stimulated thermogenesis by activating the creatine metabolism-related thermogenic pathway. Moreover, BC activated β-carotene oxygenase 1 (BCO1), which efficiently cleaved BC to retinal and consequently converted to its transcriptionally active form retinoic acid. These BC conversion products also exhibited thermogenic effects comparable to a similar level of BC. The mechanistic study revealed that retinal exhibited thermogenic activity independently of retinoic acid and retinoic acid-mediated thermogenesis was resulted partly from conversion of retinal. Moreover, BC activated α1-AR and UCP1-independent thermogenic effectors independently of UCP1 expression. In conclusion, the thermogenic response to BC and its conversion products in 3T3-L1 white adipocytes involves two interacting pathways, one mediated via β3-adrenergic receptors (β3-AR) and cyclic adenosine monophosphate (cAMP) and the other via α1-AR and increases in cytosolic Ca2+ levels activated by calcium regulatory proteins.
Collapse
Affiliation(s)
- Minji Choi
- 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
|
35
|
Jiang J, Zhou D, Zhang A, Yu W, Du L, Yuan H, Zhang C, Wang Z, Jia X, Zhang ZN, Luan B. Thermogenic adipocyte-derived zinc promotes sympathetic innervation in male mice. Nat Metab 2023; 5:481-494. [PMID: 36879120 DOI: 10.1038/s42255-023-00751-9] [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: 01/07/2022] [Accepted: 01/31/2023] [Indexed: 03/08/2023]
Abstract
Sympathetic neurons activate thermogenic adipocytes through release of catecholamine; however, the regulation of sympathetic innervation by thermogenic adipocytes is unclear. Here, we identify primary zinc ion (Zn) as a thermogenic adipocyte-secreted factor that promotes sympathetic innervation and thermogenesis in brown adipose tissue and subcutaneous white adipose tissue in male mice. Depleting thermogenic adipocytes or antagonizing β3-adrenergic receptor on adipocytes impairs sympathetic innervation. In obesity, inflammation-induced upregulation of Zn chaperone protein metallothionein-2 decreases Zn secretion from thermogenic adipocytes and leads to decreased energy expenditure. Furthermore, Zn supplementation ameliorates obesity by promoting sympathetic neuron-induced thermogenesis, while sympathetic denervation abrogates this antiobesity effect. Thus, we have identified a positive feedback mechanism for the reciprocal regulation of thermogenic adipocytes and sympathetic neurons. This mechanism is important for adaptive thermogenesis and could serve as a potential target for the treatment of obesity.
Collapse
Affiliation(s)
- Junkun Jiang
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Donglei Zhou
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Anke Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenjing Yu
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Lei Du
- Department of Metabolic Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huiwen Yuan
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Chuan Zhang
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Zelin Wang
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Xuyang Jia
- Department of Metabolic Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhen-Ning Zhang
- Translational Medical Center for Stem Cell Therapy & Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Bing Luan
- Department of Endocrinology, Tongji Hospital Affiliated to Tongji University School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
36
|
Zhu L, Tang Y, Li XY, Kerk SA, Lyssiotis CA, Feng W, Sun X, Hespe GE, Wang Z, Stemmler MP, Brabletz S, Brabletz T, Keller ET, Ma J, Cho JS, Yang J, Weiss SJ. A Zeb1/MtCK1 metabolic axis controls osteoclast activation and skeletal remodeling. EMBO J 2023; 42:e111148. [PMID: 36843552 PMCID: PMC10068323 DOI: 10.15252/embj.2022111148] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/28/2023] Open
Abstract
Osteoclasts are bone-resorbing polykaryons responsible for skeletal remodeling during health and disease. Coincident with their differentiation from myeloid precursors, osteoclasts undergo extensive transcriptional and metabolic reprogramming in order to acquire the cellular machinery necessary to demineralize bone and digest its interwoven extracellular matrix. While attempting to identify new regulatory molecules critical to bone resorption, we discovered that murine and human osteoclast differentiation is accompanied by the expression of Zeb1, a zinc-finger transcriptional repressor whose role in normal development is most frequently linked to the control of epithelial-mesenchymal programs. However, following targeting, we find that Zeb1 serves as an unexpected regulator of osteoclast energy metabolism. In vivo, Zeb1-null osteoclasts assume a hyperactivated state, markedly decreasing bone density due to excessive resorptive activity. Mechanistically, Zeb1 acts in a rheostat-like fashion to modulate murine and human osteoclast activity by transcriptionally repressing an ATP-buffering enzyme, mitochondrial creatine kinase 1 (MtCK1), thereby controlling the phosphocreatine energy shuttle and mitochondrial respiration. Together, these studies identify a novel Zeb1/MtCK1 axis that exerts metabolic control over bone resorption in vitro and in vivo.
Collapse
Affiliation(s)
- Lingxin Zhu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Yi Tang
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Xiao-Yan Li
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Samuel A Kerk
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA.,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Costas A Lyssiotis
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Wenqing Feng
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Xiaoyue Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Geoffrey E Hespe
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.,Department of Surgery, Section of Plastic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Zijun Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Marc P Stemmler
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, FAU University Erlangen-Nürnberg, Erlangen, Germany
| | - Simone Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, FAU University Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, FAU University Erlangen-Nürnberg, Erlangen, Germany
| | - Evan T Keller
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.,Department of Urology and the Institute of Gerontology, University of Michigan, Ann Arbor, MI, USA
| | - Jun Ma
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jung-Sun Cho
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jingwen Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Stephen J Weiss
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
37
|
Chen Y, Jiang Y, Cui T, Hou L, Zhao R, Bo S, Zou L, Yin C. Creatine ameliorates high-fat diet-induced obesity by regulation of lipolysis and lipophagy in brown adipose tissue and liver. Biochimie 2023; 209:85-94. [PMID: 36773834 DOI: 10.1016/j.biochi.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 01/20/2023] [Accepted: 02/08/2023] [Indexed: 02/11/2023]
Abstract
Diet-induced obesity in mice and humans is commonly associated with an imbalance between energy intake and expenditure. Reportedly, creatine can enhance energy expenditure in brown adipose tissue and reduce hepatic triglycerides accumulation; however, the molecular mechanism underlying the role of exogenous creatine supplementation in regulating lipid droplet mobilization remains elusive. Herein, we employed a high-fat diet (HFD)- induced mouse model to investigate the role of creatine in regulating lipolysis and lipophagy in brown adipose tissue and the liver. Exogenous creatine supplementation ameliorated HFD-induced obesity, increased insulin sensitivity and improved glucose homeostasis. Creatine supplementation enhanced the expression of uncoupling protein 1 (UCP1), cell death-inducing DNA fragmentation factor alpha-like effector A (CIDEA), and other brown adipose tissue-specific thermogenic genes Cpt1a, Gyk, and Pgc1β in brown adipose tissue. Furthermore, creatine inhibited the expression of CIDEA, which promotes hepatic lipid accumulation. Creatine stimulated the expression of triglyceride lipase adipose triglyceride lipase, and phospho-hormone-sensitive lipase (HSL) induced increased lipolysis in brown adipose tissue and the liver. Meanwhile, reduced LC3B expression was accompanied by an increased level of p62 in HFD-fed mice, indicating diminished basal autophagy in brown adipose tissue and the liver; however, creatine enhanced P62/LC3B induced lipophagy in brown adipose tissue and the liver. Collectively, our results suggest that creatine may function as a brown adipose tissue activator to increase whole-body energy metabolism via coordinated lipolysis and lipophagy in brown adipose tissue and the liver.
Collapse
Affiliation(s)
- Yongjie Chen
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China.
| | - Yanbin Jiang
- College of Kinesiology and Health, Capital University of Physical Education and Sports, Beijing, China
| | - Tianyu Cui
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Lei Hou
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Rong Zhao
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Shumin Bo
- College of Kinesiology and Health, Capital University of Physical Education and Sports, Beijing, China.
| | - Liying Zou
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China.
| | - Chenghong Yin
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China.
| |
Collapse
|
38
|
Haddish K, Yun JW. Dopaminergic and adrenergic receptors synergistically stimulate browning in 3T3-L1 white adipocytes. J Physiol Biochem 2023; 79:117-131. [PMID: 36342617 DOI: 10.1007/s13105-022-00928-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 10/19/2022] [Indexed: 11/09/2022]
Abstract
The browning of white adipose tissue (WAT) has attracted considerable attention in the scientific community as a popular strategy for enhancing energy expenditure to combat obesity. As a part of this strategy, β3-adrenergic receptor (β3-AR) is the most widely studied receptor that mediates thermogenesis. Parenthetically, further studies in search for additional receptors expressed in adipocytes that can mediate thermogenesis has been appearing, and this paper reports that dopaminergic receptor 1 (DRD1) and β3-AR synergistically stimulate browning in 3T3-L1 white adipocytes. qRT-PCR and immunoblot analysis methods were applied to evaluate the effects of DRD1 on the target proteins downstream of β3-AR and other markers involved in lipid metabolism, mitochondrial biogenesis, and browning events. These results show that DRD1 is expressed in epididymal WAT (eWAT), brown adipose tissue (BAT), and inguinal WAT (iWAT) of normal and high-fat-fed mice, and a deficiency of DRD1 downregulates the expression of brown adipocyte-specific proteins. Silencing of DRD1 affected lipid metabolic activity in 3T3-L1 adipocytes by reducing mitochondrial biogenesis as well as levels of lipolytic and fat oxidative marker proteins in a similar pattern to β3-AR. Moreover, mechanistic studies showed that the depletion of DRD1 downregulates β3-AR and its downstream molecules, suggesting both receptors might synergistically stimulate browning. Parallel to the UCP1-dependent thermogenesis, the depletion of DRD1 also downregulates the expression of core proteins responsible for UCP1-independent thermogenesis. Overall, DRD1 mediates β3-AR-dependent 3T3-L1 browning and UCP1-independent thermogenesis.
Collapse
Affiliation(s)
- Kiros Haddish
- 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
|
39
|
Thermogenic Adipose Redox Mechanisms: Potential Targets for Metabolic Disease Therapies. Antioxidants (Basel) 2023; 12:antiox12010196. [PMID: 36671058 PMCID: PMC9854447 DOI: 10.3390/antiox12010196] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/07/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Metabolic diseases, such as diabetes and non-alcoholic fatty liver disease (NAFLD), have several negative health outcomes on affected humans. Dysregulated energy metabolism is a key component underlying the pathophysiology of these conditions. Adipose tissue is a fundamental regulator of energy homeostasis that utilizes several redox reactions to carry out the metabolism. Brown and beige adipose tissues, in particular, perform highly oxidative reactions during non-shivering thermogenesis to dissipate energy as heat. The appropriate regulation of energy metabolism then requires coordinated antioxidant mechanisms to counterbalance the oxidation reactions. Indeed, non-shivering thermogenesis activation can cause striking changes in concentrations of both oxidants and antioxidants in order to adapt to various oxidative environments. Current therapeutic options for metabolic diseases either translate poorly from rodent models to humans (in part due to the challenges of creating a physiologically relevant rodent model) or tend to have numerous side effects, necessitating novel therapies. As increased brown adipose tissue activity results in enhanced energy expenditure and is associated with beneficial effects on metabolic health, such as decreased obesity, it has gathered great interest as a modulator of metabolic disease. One potential reason for the beneficial health effects may be that although non-shivering thermogenesis is enormously oxidative, it is also associated with decreased oxidant formation after its activation. However, targeting its redox mechanisms specifically to alter metabolic disease remains an underexplored area. Therefore, this review will discuss the role of adipose tissue in energy homeostasis, non-shivering thermogenesis in adults, and redox mechanisms that may serve as novel therapeutic targets of metabolic disease.
Collapse
|
40
|
Curcumin Stimulates UCP1-independent Thermogenesis in 3T3-L1 White Adipocytes but Suppresses in C2C12 Muscle Cells. BIOTECHNOL BIOPROC E 2022. [DOI: 10.1007/s12257-022-0319-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
41
|
Daneshyar S, Ghasemnian A, Mirakhori Z, Daneshyar S. The effect of high fat diet and endurance training on newly discovery of nonshivering-thermogenic factors under thermoneutrality in mice. Sci Sports 2022. [DOI: 10.1016/j.scispo.2022.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
42
|
Selenium and selenoproteins in thermogenic adipocytes. Arch Biochem Biophys 2022; 731:109445. [PMID: 36265651 PMCID: PMC9981474 DOI: 10.1016/j.abb.2022.109445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/20/2022]
Abstract
Selenium (Se) is involved in energy metabolism in the liver, white adipose tissue, and skeletal muscle, and may also play a role in thermogenic adipocytes, i.e. brown and beige adipocytes. Thereby this micronutrient is a key nutritional target to aid in combating obesity and metabolic diseases. In thermogenic adipocytes, particularly in brown adipose tissue (BAT), the selenoprotein type 2 iodothyronine deiodinase (DIO2) is essential for the activation of adaptive thermogenesis. Recent evidence has suggested that additional selenoproteins may also be participating in this process, and a role for Se itself through its metabolic pathways is also envisioned. In this review, we discuss the recognized effects and the knowledge gaps in the involvement of Se metabolism and selenoproteins in the mechanisms of adaptive thermogenesis in thermogenic (brown and beige) adipocytes.
Collapse
|
43
|
AlZaim I, Eid AH, Abd-Elrahman KS, El-Yazbi AF. Adipose Tissue Mitochondrial Dysfunction and Cardiometabolic Diseases: On the Search for Novel Molecular Targets. Biochem Pharmacol 2022; 206:115337. [DOI: 10.1016/j.bcp.2022.115337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/17/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
|
44
|
Abstract
Synchronizing food intake with the body clock boosts thermogenesis and limits obesity.
Collapse
Affiliation(s)
- Damien Lagarde
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Lawrence Kazak
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| |
Collapse
|
45
|
Hepler C, Weidemann BJ, Waldeck NJ, Marcheva B, Cedernaes J, Thorne AK, Kobayashi Y, Nozawa R, Newman MV, Gao P, Shao M, Ramsey KM, Gupta RK, Bass J. Time-restricted feeding mitigates obesity through adipocyte thermogenesis. Science 2022; 378:276-284. [PMID: 36264811 PMCID: PMC10150371 DOI: 10.1126/science.abl8007] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Misalignment of feeding rhythms with the light-dark cycle leads to disrupted peripheral circadian clocks and obesity. Conversely, restricting feeding to the active period mitigates metabolic syndrome through mechanisms that remain unknown. We found that genetic enhancement of adipocyte thermogenesis through ablation of the zinc finger protein 423 (ZFP423) attenuated obesity caused by consumption of a high-fat diet during the inactive (light) period by increasing futile creatine cycling in mice. Circadian control of adipocyte creatine metabolism underlies the timing of diet-induced thermogenesis, and enhancement of adipocyte circadian rhythms through overexpression of the clock activator brain and muscle Arnt-like protein-1 (BMAL1) ameliorated metabolic complications during diet-induced obesity. These findings uncover rhythmic creatine-mediated thermogenesis as an essential mechanism that drives metabolic benefits during time-restricted feeding.
Collapse
Affiliation(s)
- Chelsea Hepler
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Benjamin J. Weidemann
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Nathan J. Waldeck
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Biliana Marcheva
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jonathan Cedernaes
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Anneke K. Thorne
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yumiko Kobayashi
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Rino Nozawa
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Marsha V. Newman
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Peng Gao
- Robert H. Lurie Cancer Center Metabolomics Core, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Mengle Shao
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Kathryn M. Ramsey
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Rana K. Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Joseph Bass
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
46
|
Beignon F, Gueguen N, Tricoire-Leignel H, Mattei C, Lenaers G. The multiple facets of mitochondrial regulations controlling cellular thermogenesis. Cell Mol Life Sci 2022; 79:525. [PMID: 36125552 DOI: 10.1007/s00018-022-04523-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/21/2022] [Accepted: 08/09/2022] [Indexed: 12/01/2022]
Abstract
Understanding temperature production and regulation in endotherm organisms becomes a crucial challenge facing the increased frequency and intensity of heat strokes related to global warming. Mitochondria, located at the crossroad of metabolism, respiration, Ca2+ homeostasis, and apoptosis, were recently proposed to further act as cellular radiators, with an estimated inner temperature reaching 50 °C in common cell lines. This inner thermogenesis might be further exacerbated in organs devoted to produce consistent efforts as muscles, or heat as brown adipose tissue, in response to acute solicitations. Consequently, pathways promoting respiratory chain uncoupling and mitochondrial activity, such as Ca2+ fluxes, uncoupling proteins, futile cycling, and substrate supplies, provide the main processes controlling heat production and cell temperature. The mitochondrial thermogenesis might be further amplified by cytoplasmic mechanisms promoting the over-consumption of ATP pools. Considering these new thermic paradigms, we discuss here all conventional wisdoms linking mitochondrial functions to cellular thermogenesis in different physiological conditions.
Collapse
Affiliation(s)
- Florian Beignon
- Univ Angers, MitoLab, Unité MITOVASC, UMR CNRS 6015, INSERM U1083, SFR ICAT, Angers, France.
| | - Naig Gueguen
- Univ Angers, MitoLab, Unité MITOVASC, UMR CNRS 6015, INSERM U1083, SFR ICAT, Angers, France.,Service de Biochimie et Biologie Moléculaire, CHU d'Angers, Angers, France
| | | | - César Mattei
- Univ Angers, CarMe, Unité MITOVASC, UMR CNRS 6015, INSERM U1083, SFR ICAT, Angers, France
| | - Guy Lenaers
- Univ Angers, MitoLab, Unité MITOVASC, UMR CNRS 6015, INSERM U1083, SFR ICAT, Angers, France. .,Service de Neurologie, CHU d'Angers, Angers, France.
| |
Collapse
|
47
|
Yin X, Chen Y, Ruze R, Xu R, Song J, Wang C, Xu Q. The evolving view of thermogenic fat and its implications in cancer and metabolic diseases. Signal Transduct Target Ther 2022; 7:324. [PMID: 36114195 PMCID: PMC9481605 DOI: 10.1038/s41392-022-01178-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 02/07/2023] Open
Abstract
AbstractThe incidence of metabolism-related diseases like obesity and type 2 diabetes mellitus has reached pandemic levels worldwide and increased gradually. Most of them are listed on the table of high-risk factors for malignancy, and metabolic disorders systematically or locally contribute to cancer progression and poor prognosis of patients. Importantly, adipose tissue is fundamental to the occurrence and development of these metabolic disorders. White adipose tissue stores excessive energy, while thermogenic fat including brown and beige adipose tissue dissipates energy to generate heat. In addition to thermogenesis, beige and brown adipocytes also function as dynamic secretory cells and a metabolic sink of nutrients, like glucose, fatty acids, and amino acids. Accordingly, strategies that activate and expand thermogenic adipose tissue offer therapeutic promise to combat overweight, diabetes, and other metabolic disorders through increasing energy expenditure and enhancing glucose tolerance. With a better understanding of its origins and biological functions and the advances in imaging techniques detecting thermogenesis, the roles of thermogenic adipose tissue in tumors have been revealed gradually. On the one hand, enhanced browning of subcutaneous fatty tissue results in weight loss and cancer-associated cachexia. On the other hand, locally activated thermogenic adipocytes in the tumor microenvironment accelerate cancer progression by offering fuel sources and is likely to develop resistance to chemotherapy. Here, we enumerate current knowledge about the significant advances made in the origin and physiological functions of thermogenic fat. In addition, we discuss the multiple roles of thermogenic adipocytes in different tumors. Ultimately, we summarize imaging technologies for identifying thermogenic adipose tissue and pharmacologic agents via modulating thermogenesis in preclinical experiments and clinical trials.
Collapse
|
48
|
Weber A, Medak KD, Townsend LK, Wright DC. Ketogenic diet induced weight loss occurs independent of housing temperature and is followed by hyperphagia and weight regain after cessation in mice. J Physiol 2022; 600:4677-4693. [PMID: 36083198 DOI: 10.1113/jp283469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/24/2022] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Ketogenic diets reduce food intake, increase energy expenditure and cause weight loss in rodents Prior preclinical studies have been completed at room temperature, a condition which induces thermal stress and limits clinical translatability We demonstrate that ketogenic diet-induced reductions in food intake, increases in energy expenditure, weight loss and improvements in glucose homeostasis are similar in mice housed at room temperature or thermal neutrality Ketogenic diet induced reductions in food intake appear to explain a large degree of weight loss. Similarly, switching mice from a ketogenic to an obesogenic diet leads to hyperphagia mediated weight gain ABSTRACT: Ketogenic diets (KDs) are a popular tool used for weight management. Studies in mice have demonstrated that KDs reduce food intake, increase energy expenditure and cause weight loss. These studies were completed at room temperature (RT), a condition below the animal's thermal neutral (TN) zone which induces thermal stress. As energy intake and expenditure are sensitive to environmental temperature it's not clear if a KD would exert the same beneficial effects under TN conditions. Adherence to restrictive diets is poor and consequently it is important to examine the effects, and underlying mechanisms, of cycling from a ketogenic to an obesogenic diet. The purpose of the current study was to determine if housing temperature impacted the effects of a KD in obese mice and to determine if the mechanisms driving KD-induced weight loss reverse when mice are switched to an obesogenic high fat diet. We demonstrate that KD-induced reductions in food intake, increases in energy expenditure, weight loss and improvements in glucose homeostasis are not dependent upon housing temperature. KD-induced weight loss, seems to be largely explained by reductions in caloric intake while cycling mice back to an obesogenic diet following a period of KD feeding leads to hyperphagia-induced weight gain. Collectively, our results suggest that prior findings with mice fed a KD at RT are likely not an artifact of how mice were housed and that initial changes in weight when transitioning from an obesogenic to a ketogenic diet or back, are largely dependent on food intake. Abstract figure legend The impact of housing temperature on ketogenic diet mediated changes in energy expenditure, food intake and weight gain. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Alyssa Weber
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Kyle D Medak
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Logan K Townsend
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - David C Wright
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada.,Faculty of Land and Food Systems, University of British Columbia, Vancouver, British Columbia, Canada.,British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
49
|
Meneses MJ, Sousa-Lima I, Jarak I, Raposo JF, Alves MG, Macedo MP. Distinct impacts of fat and fructose on the liver, muscle, and adipose tissue metabolome: An integrated view. Front Endocrinol (Lausanne) 2022; 13:898471. [PMID: 36060961 PMCID: PMC9428722 DOI: 10.3389/fendo.2022.898471] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022] Open
Abstract
Objective In the last years, changes in dietary habits have contributed to the increasing prevalence of metabolic disorders, such as non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM). The differential burden of lipids and fructose on distinct organs needs to be unveiled. Herein, we hypothesized that high-fat and high-fructose diets differentially affect the metabolome of insulin-sensitive organs such as the liver, muscle, and different adipose tissue depots. Methods We have studied the impact of 12 weeks of a control (11.50% calories from fat, 26.93% from protein, and 61.57% from carbohydrates), high-fat/sucrose (HFat), or high-fructose (HFruct) feeding on C57Bl/6J male mice. Besides glucose homeostasis, we analyzed the hepatic levels of glucose and lipid-metabolism-related genes and the metabolome of the liver, the muscle, and white (WAT) and brown adipose tissue (BAT) depots. Results HFat diet led to a more profound impact on hepatic glucose and lipid metabolism than HFruct, with mice presenting glucose intolerance, increased saturated fatty acids, and no glycogen pool, yet both HFat and HFruct presented hepatic insulin resistance. HFat diet promoted a decrease in glucose and lactate pools in the muscle and an increase in glutamate levels. While HFat had alterations in BAT metabolites that indicate increased thermogenesis, HFruct led to an increase in betaine, a protective metabolite against fructose-induced inflammation. Conclusions Our data illustrate that HFat and HFruct have a negative but distinct impact on the metabolome of the liver, muscle, WAT, and BAT.
Collapse
Affiliation(s)
- Maria João Meneses
- iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas (NMS/FCM), Universidade Nova de Lisboa, Lisbon, Portugal
- Portuguese Diabetes Association - Education and Research Center (APDP-ERC), Lisbon, Portugal
| | - Inês Sousa-Lima
- iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas (NMS/FCM), Universidade Nova de Lisboa, Lisbon, Portugal
| | - Ivana Jarak
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - João F. Raposo
- iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas (NMS/FCM), Universidade Nova de Lisboa, Lisbon, Portugal
- Portuguese Diabetes Association - Education and Research Center (APDP-ERC), Lisbon, Portugal
| | - Marco G. Alves
- Department of Anatomy and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Maria Paula Macedo
- iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas (NMS/FCM), Universidade Nova de Lisboa, Lisbon, Portugal
- Portuguese Diabetes Association - Education and Research Center (APDP-ERC), Lisbon, Portugal
- Medical Sciences Department, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
50
|
Lygate CA, Lake HA, McAndrew DJ, Neubauer S, Zervou S. Influence of homoarginine on creatine accumulation and biosynthesis in the mouse. Front Nutr 2022; 9:969702. [PMID: 36017222 PMCID: PMC9395972 DOI: 10.3389/fnut.2022.969702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/21/2022] [Indexed: 12/24/2022] Open
Abstract
Organisms obtain creatine from their diet or by de novo synthesis via AGAT (L-arginine:glycine amidinotransferase) and GAMT (Guanidinoacetate N-methyltrasferase) in kidney and liver, respectively. AGAT also synthesizes homoarginine (hArg), low levels of which predict poor outcomes in human cardiovascular disease, while supplementation maintains contractility in murine heart failure. However, the expression pattern of AGAT has not been systematically studied in mouse tissues and nothing is known about potential feedback interactions between creatine and hArg. Herein, we show that C57BL/6J mice express AGAT and GAMT in kidney and liver respectively, whereas pancreas was the only organ to express appreciable levels of both enzymes, but no detectable transmembrane creatine transporter (Slc6A8). In contrast, kidney, left ventricle (LV), skeletal muscle and brown adipose tissue must rely on creatine transporter for uptake, since biosynthetic enzymes are not expressed. The effects of creatine and hArg supplementation were then tested in wild-type and AGAT knockout mice. Homoarginine did not alter creatine accumulation in plasma, LV or kidney, whereas in pancreas from AGAT KO, the addition of hArg resulted in higher levels of tissue creatine than creatine-supplementation alone (P < 0.05). AGAT protein expression in kidney was downregulated by creatine supplementation (P < 0.05), consistent with previous reports of end-product repression. For the first time, we show that hArg supplementation causes a similar down-regulation of AGAT protein (P < 0.05). These effects on AGAT were absent in the pancreas, suggesting organ specific mechanisms of regulation. These findings highlight the potential for interactions between creatine and hArg that may have implications for the use of dietary supplements and other therapeutic interventions.
Collapse
Affiliation(s)
- Craig A. Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, United Kingdom
| | | | | | | | - Sevasti Zervou
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre for Research Excellence, University of Oxford, Oxford, United Kingdom
| |
Collapse
|