1
|
Zhu K, Liu S, Huang Y, Zhang B, Houssein N, Wu J. Chrna2-driven CRE Is Expressed in Beige Adipocytes. Endocrinology 2024; 166:bqae153. [PMID: 39540707 PMCID: PMC11630559 DOI: 10.1210/endocr/bqae153] [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: 07/15/2024] [Revised: 10/30/2024] [Accepted: 11/13/2024] [Indexed: 11/16/2024]
Abstract
Significant research interest has been focused on beige adipocytes, the activation of which improves glucose and lipid homeostasis, therefore representing new therapeutic opportunities for metabolic diseases. Various Cre/Lox-based strategies have been used to investigate the developmental history of beige adipocytes and how these cells adapt to environmental changes. Despite the significant advancement of our understanding of beige adipocyte biology, much of the molecular insights of the beige adipocyte, including its origin and cell type-specific function, remain to be further illustrated. It has previously been shown that Chrna2 (cholinergic receptor nicotinic alpha 2 subunit) has selective functionality in beige adipocytes. In this study, we explore the Chrna2-Cre-driven reporter expression in mouse beige adipocytes in vivo and in vitro. Our findings indicate that Chrna2-Cre expression is present selectively in multiple locular beige adipocytes in subcutaneous inguinal white adipose tissue (iWAT) and differentiated stromal vascular fraction from iWAT. Chrna2-Cre expression was detected in iWAT of young pups and mice after cold exposure where a significant number of beige adipocytes are present. Chrna2-Cre-driven reporter expression is permanent in iWAT postlabeling and can be detected in the iWAT of adult mice or mice that have been housed extensively at thermoneutrality after cold exposure, even though only "inactive dormant" beige adipocytes are present in these mice. Chrna2-Cre expression can also be increased by rosiglitazone treatment and β-adrenergic activation. This research, therefore, introduces the Chrna2-Cre line as a valuable tool for tracking the development of beige adipocytes and investigating beige fat function.
Collapse
Affiliation(s)
- Kezhou Zhu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shanshan Liu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yunying Huang
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Biyang Zhang
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nadia Houssein
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jun Wu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
2
|
Estrada-Meza J, Videlo J, Bron C, Duchampt A, Saint-Béat C, Zergane M, Silva M, Rajas F, Bouret SG, Mithieux G, Gautier-Stein A. Intestinal gluconeogenesis controls the neonatal development of hypothalamic feeding circuits. Mol Metab 2024; 89:102036. [PMID: 39304064 PMCID: PMC11470480 DOI: 10.1016/j.molmet.2024.102036] [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: 07/13/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024] Open
Abstract
OBJECTIVE Intestinal gluconeogenesis (IGN) regulates adult energy homeostasis in part by controlling the same hypothalamic targets as leptin. In neonates, leptin exhibits a neonatal surge controlling axonal outgrowth between the different hypothalamic nuclei involved in feeding circuits and autonomic innervation of peripheral tissues involved in energy and glucose homeostasis. Interestingly, IGN is induced during this specific time-window. We hypothesized that the neonatal pic of IGN also regulates the development of hypothalamic feeding circuits and sympathetic innervation of adipose tissues. METHODS We genetically induced neonatal IGN by overexpressing G6pc1 the catalytic subunit of glucose-6-phosphatase (the mandatory enzyme of IGN) at birth or at twelve days after birth. The neonatal development of hypothalamic feeding circuits was studied by measuring Agouti-related protein (AgRP) and Pro-opiomelanocortin (POMC) fiber density in hypothalamic nuclei of 20-day-old pups. The effect of the neonatal induction of intestinal G6pc1 on sympathetic innervation of the adipose tissues was studied via tyrosine hydroxylase (TH) quantification. The metabolic consequences of the neonatal induction of intestinal G6pc1 were studied in adult mice challenged with a high-fat/high-sucrose (HFHS) diet for 2 months. RESULTS Induction of intestinal G6pc1 at birth caused a neonatal reorganization of AgRP and POMC fiber density in the paraventricular nucleus of the hypothalamus, increased brown adipose tissue tyrosine hydroxylase levels, and protected against high-fat feeding-induced metabolic disorders. In contrast, inducing intestinal G6pc1 12 days after birth did not impact AgRP/POMC fiber densities, adipose tissue innervation or adult metabolism. CONCLUSION These findings reveal that IGN at birth but not later during postnatal life controls the development of hypothalamic feeding circuits and sympathetic innervation of adipose tissues, promoting a better management of metabolism in adulthood.
Collapse
Affiliation(s)
| | - Jasmine Videlo
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Clara Bron
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Adeline Duchampt
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | | | - Mickael Zergane
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Marine Silva
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Fabienne Rajas
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Sebastien G Bouret
- University Lille, Inserm, CHU Lille, Laboratory of development and plasticity of the Neuroendocrine brain, Lille Neuroscience & Cognition, Inserm UMR-S1172, Lille, France
| | - Gilles Mithieux
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | | |
Collapse
|
3
|
Hu L, Zhang G, Tong X, Wang L, Qiu X, Yang H, Liu X, Huang H. Characterization of a novel adipose tissue located between abdominal lymph nodes and cervix/prostate in mice. Am J Physiol Endocrinol Metab 2024; 327:E134-E144. [PMID: 38747899 DOI: 10.1152/ajpendo.00110.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/15/2024] [Accepted: 05/01/2024] [Indexed: 07/18/2024]
Abstract
Perigonadal adipose tissue is a homogeneous white adipose tissue (WAT) in adult male mice without any brown adipose tissue (BAT). However, there are congenital differences in the gonads between male and female mice. Whether heterogeneity existed in perigonadal adipose tissues (ATs) in female mice remains unknown. This study reported a perigonadal brown-like AT located between abdominal lymph nodes and the uterine cervix in female mice, termed lymph node-cervical adipose tissue (LNCAT). Its counterpart, lymph node-prostatic adipose tissue (LNPAT), exhibited white phenotype in adult virgin male mice. When exposed to cold, LNCAT/LNPAT increased uncoupling protein 1 (UCP1) expression via activation of tyrosine hydroxylase (TH), in which abdominal lymph nodes were involved. Interestingly, the UCP1 expression in LNCAT/LNPAT varied under different reproductive stages. The UCP1 expression in LNCAT was upregulated at early pregnancy, declined at midlate pregnancy, and reverted in weaning dams. Mating behavior stimulated LNPAT browning in male mice. We found that androgen but not estrogen or progesterone inhibited UCP1 expression in LNCAT. Androgen administration reversed the castration-induced LNPAT browning. Our results identified a perigonadal brown-like AT in female mice and characterized its UCP1 expression patterns under various conditions.NEW & NOTEWORTHY A novel perigonadal brown-like AT (LNCAT) of female mice was identified. Abdominal lymph nodes were involved in cold-induced browning in this newly discovered adipose tissue. The UCP1 expression in LNCAT/LNPAT was also related to ages, sexes, and reproductive stages, in which androgen acted as an inhibitor role.
Collapse
Affiliation(s)
- Lihao Hu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, People's Republic of China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China
| | - Gaochen Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, People's Republic of China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China
| | - Xiaoyu Tong
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institute of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, People's Republic of China
| | - Lulu Wang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, People's Republic of China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China
| | - Xiang Qiu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, People's Republic of China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China
| | - Hongbo Yang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, People's Republic of China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China
| | - Xinmei Liu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, People's Republic of China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China
| | - Hefeng Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, People's Republic of China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, People's Republic of China
- Key Laboratory of Reproductive Genetics (Ministry of Education), Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| |
Collapse
|
4
|
Yuan Y, Hu R, Park J, Xiong S, Wang Z, Qian Y, Shi Z, Wu R, Han Z, Ong SG, Lin S, Varady KA, Xu P, Berry DC, Shu G, Jiang Y. Macrophage-derived chemokine CCL22 establishes local LN-mediated adaptive thermogenesis and energy expenditure. SCIENCE ADVANCES 2024; 10:eadn5229. [PMID: 38924414 PMCID: PMC11204298 DOI: 10.1126/sciadv.adn5229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/20/2024] [Indexed: 06/28/2024]
Abstract
There is a regional preference around lymph nodes (LNs) for adipose beiging. Here, we show that local LN removal within inguinal white adipose tissue (iWAT) greatly impairs cold-induced beiging, and this impairment can be restored by injecting M2 macrophages or macrophage-derived C-C motif chemokine (CCL22) into iWAT. CCL22 injection into iWAT effectively promotes iWAT beiging, while blocking CCL22 with antibodies can prevent it. Mechanistically, the CCL22 receptor, C-C motif chemokine receptor 4 (CCR4), within eosinophils and its downstream focal adhesion kinase/p65/interleukin-4 signaling are essential for CCL22-mediated beige adipocyte formation. Moreover, CCL22 levels are inversely correlated with body weight and fat mass in mice and humans. Acute elevation of CCL22 levels effectively prevents diet-induced body weight and fat gain by enhancing adipose beiging. Together, our data identify the CCL22-CCR4 axis as an essential mediator for LN-controlled adaptive thermogenesis and highlight its potential to combat obesity and its associated complications.
Collapse
Affiliation(s)
- Yexian Yuan
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Ruoci Hu
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jooman Park
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Shaolei Xiong
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Zilai Wang
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Yanyu Qian
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Zuoxiao Shi
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Ruifan Wu
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Zhenbo Han
- Department of Pharmacology and Regenerative Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Sang-Ging Ong
- Department of Pharmacology and Regenerative Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Shuhao Lin
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Krista A. Varady
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Pingwen Xu
- Division of Endocrinology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Daniel C. Berry
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Gang Shu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yuwei Jiang
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
- Division of Endocrinology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
5
|
Mooli RGR, Zhu B, Khan SR, Nagati V, Michealraj KA, Jurczak MJ, Ramakrishnan SK. Epigenetically active chromatin in neonatal iWAT reveals GABPα as a potential regulator of beige adipogenesis. Front Endocrinol (Lausanne) 2024; 15:1385811. [PMID: 38765953 PMCID: PMC11099907 DOI: 10.3389/fendo.2024.1385811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/12/2024] [Indexed: 05/22/2024] Open
Abstract
Background Thermogenic beige adipocytes, which dissipate energy as heat, are found in neonates and adults. Recent studies show that neonatal beige adipocytes are highly plastic and contribute to >50% of beige adipocytes in adults. Neonatal beige adipocytes are distinct from recruited beige adipocytes in that they develop independently of temperature and sympathetic innervation through poorly defined mechanisms. Methods We characterized the neonatal beige adipocytes in the inguinal white adipose tissue (iWAT) of C57BL6 postnatal day 3 and 20 mice (P3 and P20) by imaging, genome-wide RNA-seq analysis, ChIP-seq analysis, qRT-PCR validation, and biochemical assays. Results We found an increase in acetylated histone 3 lysine 27 (H3K27ac) on the promoter and enhancer regions of beige-specific gene UCP1 in iWAT of P20 mice. Furthermore, H3K27ac ChIP-seq analysis in the iWAT of P3 and P20 mice revealed strong H3K27ac signals at beige adipocyte-associated genes in the iWAT of P20 mice. The integration of H3K27ac ChIP-seq and RNA-seq analysis in the iWAT of P20 mice reveal epigenetically active signatures of beige adipocytes, including oxidative phosphorylation and mitochondrial metabolism. We identify the enrichment of GA-binding protein alpha (GABPα) binding regions in the epigenetically active chromatin regions of the P20 iWAT, particularly on beige genes, and demonstrate that GABPα is required for beige adipocyte differentiation. Moreover, transcriptomic analysis and glucose oxidation assays revealed increased glycolytic activity in the neonatal iWAT from P20. Conclusions Our findings demonstrate that epigenetic mechanisms regulate the development of peri-weaning beige adipocytes via GABPα. Further studies to better understand the upstream mechanisms that regulate epigenetic activation of GABPα and characterization of the metabolic identity of neonatal beige adipocytes will help us harness their therapeutic potential in metabolic diseases.
Collapse
Affiliation(s)
- Raja Gopal Reddy Mooli
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, PA, United States
| | - Bokai Zhu
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, PA, United States
- Pittsburgh Liver Research Centre, University of Pittsburgh, Pittsburgh, PA, United States
- Aging Institute of University of Pittsburgh Medical Center (UPMC), University of Pittsburgh, Pittsburgh, PA, United States
| | - Saifur R. Khan
- Division of Cardiology, University of Pittsburgh, Pittsburgh, PA, United States
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
- Pittsburgh VA Medical Centre, Pittsburgh, PA, United States
- Center for Immunometabolism, University of Pittsburgh, Pittsburgh, PA, United States
| | - Veerababu Nagati
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, PA, United States
| | | | - Michael J. Jurczak
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, PA, United States
| | - Sadeesh K. Ramakrishnan
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, PA, United States
- Pittsburgh Liver Research Centre, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
6
|
de Sousa É, de Mendonça M, Bolin AP, de Oliveira NP, Real CC, Hu X, Huang ZP, Wang DZ, Rodrigues AC. Sex-specific regulation of miR-22 and ERα in white adipose tissue of obese dam's female offspring impairs the early postnatal development of functional beige adipocytes in mice. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167057. [PMID: 38331111 DOI: 10.1016/j.bbadis.2024.167057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 12/31/2023] [Accepted: 01/30/2024] [Indexed: 02/10/2024]
Abstract
During inguinal adipose tissue (iWAT) ontogenesis, beige adipocytes spontaneously appear between postnatal 10 (P10) and P20 and their ablation impairs iWAT browning capacity in adulthood. Since maternal obesity has deleterious effects on offspring iWAT function, we aimed to investigate its effect in spontaneous iWAT browning in offspring. Female C57BL/6 J mice were fed a control or obesogenic diet six weeks before mating. Male and female offspring were euthanized at P10 and P20 or weaned at P21 and fed chow diet until P60. At P50, mice were treated with saline or CL316,243, a β3-adrenoceptor agonist, for ten days. Maternal obesity induced insulin resistance at P60, and CL316,243 treatment effectively restored insulin sensitivity in male but not female offspring. This discrepancy occurred due to female offspring severe browning impairment. During development, the spontaneous iWAT browning and sympathetic nerve branching at P20 were severely impaired in female obese dam's offspring but occurred normally in males. Additionally, maternal obesity increased miR-22 expression in the iWAT of male and female offspring during development. ERα, a target and regulator of miR-22, was concomitantly upregulated in the male's iWAT. Next, we evaluated miR-22 knockout (KO) offspring at P10 and P20. The miR-22 deficiency does not affect spontaneous iWAT browning in females and, surprisingly, anticipates iWAT browning in males. In conclusion, maternal obesity impairs functional iWAT development in the offspring in a sex-specific way that seems to be driven by miR-22 levels and ERα signaling. This impacts adult browning capacity and glucose homeostasis, especially in female offspring.
Collapse
Affiliation(s)
- Érica de Sousa
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Mariana de Mendonça
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Anaysa Paola Bolin
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Nayara Preste de Oliveira
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | | - Xiaoyun Hu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhan-Peng Huang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alice Cristina Rodrigues
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
7
|
Wang Y, Ye L. The Afferent Function of Adipose Innervation. Diabetes 2024; 73:348-354. [PMID: 38377447 PMCID: PMC10882147 DOI: 10.2337/dbi23-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 12/18/2023] [Indexed: 02/22/2024]
Abstract
Adipose tissue innervation is critical for regulating metabolic and energy homeostasis. While the sympathetic efferent innervation of fat is well characterized, the role of sensory or afferent innervation remains less explored. This article reviews previous work on adipose innervation and recent advances in the study of sensory innervation of adipose tissues. We discuss key open questions, including the physiological implications of adipose afferents in homeostasis as well as potential cross talk with sympathetic neurons, the immune system, and hormonal pathways. We also outline the general technical challenges of studying dorsal root ganglia innervating fat, along with emerging technologies that may overcome these barriers. Finally, we highlight areas for further research to deepen our understanding of the afferent function of adipose innervation.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA
| | - Li Ye
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA
| |
Collapse
|
8
|
Ren W, Hua M, Cao F, Zeng W. The Sympathetic-Immune Milieu in Metabolic Health and Diseases: Insights from Pancreas, Liver, Intestine, and Adipose Tissues. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306128. [PMID: 38039489 PMCID: PMC10885671 DOI: 10.1002/advs.202306128] [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: 08/28/2023] [Revised: 10/28/2023] [Indexed: 12/03/2023]
Abstract
Sympathetic innervation plays a crucial role in maintaining energy balance and contributes to metabolic pathophysiology. Recent evidence has begun to uncover the innervation landscape of sympathetic projections and sheds light on their important functions in metabolic activities. Additionally, the immune system has long been studied for its essential roles in metabolic health and diseases. In this review, the aim is to provide an overview of the current research progress on the sympathetic regulation of key metabolic organs, including the pancreas, liver, intestine, and adipose tissues. In particular, efforts are made to highlight the critical roles of the peripheral nervous system and its potential interplay with immune components. Overall, it is hoped to underscore the importance of studying metabolic organs from a comprehensive and interconnected perspective, which will provide valuable insights into the complex mechanisms underlying metabolic regulation and may lead to novel therapeutic strategies for metabolic diseases.
Collapse
Affiliation(s)
- Wenran Ren
- Institute for Immunology and School of MedicineTsinghua Universityand Tsinghua‐Peking Center for Life SciencesBeijing100084China
| | - Meng Hua
- Institute for Immunology and School of MedicineTsinghua Universityand Tsinghua‐Peking Center for Life SciencesBeijing100084China
| | - Fang Cao
- Department of NeurosurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhou563000China
| | - Wenwen Zeng
- Institute for Immunology and School of MedicineTsinghua Universityand Tsinghua‐Peking Center for Life SciencesBeijing100084China
- SXMU‐Tsinghua Collaborative Innovation Center for Frontier MedicineTaiyuan030001China
- Beijing Key Laboratory for Immunological Research on Chronic DiseasesBeijing100084China
| |
Collapse
|
9
|
Tsukada A, Okamatsu-Ogura Y, Futagawa E, Habu Y, Takahashi N, Kato-Suzuki M, Kato Y, Ishizuka S, Sonoyama K, Kimura K. White adipose tissue undergoes browning during preweaning period in association with microbiota formation in mice. iScience 2023; 26:107239. [PMID: 37485363 PMCID: PMC10362363 DOI: 10.1016/j.isci.2023.107239] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/11/2023] [Accepted: 06/26/2023] [Indexed: 07/25/2023] Open
Abstract
Beige adipocytes are transiently induced during early postnatal period in mice. Previous studies have suggested that, unlike in adults, the induction is independent of the sympathetic nerve activity; however, the mechanism is yet unknown. Here, we showed that beige adipocytes are induced during the preweaning period in association with the formation of microbiota in mice. Alteration of gut microbiota composition in preweaning mice by maternal treatment with antibiotics or high-fat diet feeding substantially suppressed WAT browning. The suppression was also found in pups transplanted cecal microbiota from pups of high-fat diet-fed dams. These treatments reduced the hepatic expression of genes involved in bile acid synthesis and the serum bile acids level. The abundance of Porphyromonadaceae and Ruminococcaceae in microbiota showed a positive and negative correlation with the induction of beige adipocytes, respectively. This finding may provide comprehensive understanding of the association between gut microbiota and adipose tissue development in the neonatal period.
Collapse
Affiliation(s)
- Anju Tsukada
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Yuko Okamatsu-Ogura
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Emi Futagawa
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Yuki Habu
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Natsumi Takahashi
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Mira Kato-Suzuki
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Yuko Kato
- Laboratory of Nutritional Biochemistry, Research Faculty of Agriculture, Hokkaido University, Sapporo 060-0809, Japan
| | - Satoshi Ishizuka
- Laboratory of Nutritional Biochemistry, Research Faculty of Agriculture, Hokkaido University, Sapporo 060-0809, Japan
| | - Kei Sonoyama
- Laboratory of Food Biochemistry, Research Faculty of Agriculture, Hokkaido University, Sapporo 060-0809, Japan
| | - Kazuhiro Kimura
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| |
Collapse
|
10
|
Shamsi F, Zheng R, Ho LL, Chen K, Tseng YH. Comprehensive analysis of intercellular communication in the thermogenic adipose niche. Commun Biol 2023; 6:761. [PMID: 37479789 PMCID: PMC10361964 DOI: 10.1038/s42003-023-05140-2] [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: 11/21/2022] [Accepted: 07/12/2023] [Indexed: 07/23/2023] Open
Abstract
Brown adipose tissue (BAT) is responsible for regulating body temperature through adaptive thermogenesis. The ability of thermogenic adipocytes to dissipate chemical energy as heat counteracts weight gain and has gained considerable attention as a strategy against obesity. BAT undergoes major remodeling in a cold environment. This remodeling results from changes in the number and function of brown adipocytes, expanding the network of blood vessels and sympathetic nerves, and changes in the composition and function of immune cells. Such synergistic adaptation requires extensive crosstalk between individual cells in the tissue to coordinate their responses. To understand the mechanisms of intercellular communication in BAT, we apply the CellChat algorithm to single-cell transcriptomic data of mouse BAT. We construct an integrative network of the ligand-receptor interactome in BAT and identify the major signaling inputs and outputs of each cell type. By comparing the ligand-receptor interactions in BAT of mice housed at different environmental temperatures, we show that cold exposure enhances the intercellular interactions among the major cell types in BAT, including adipocytes, adipocyte progenitors, lymphatic and vascular endothelial cells, myelinated and non-myelinated Schwann cells, and immune cells. These interactions are predicted to regulate the remodeling of the extracellular matrix, the inflammatory response, angiogenesis, and neurite growth. Together, our integrative analysis of intercellular communications in BAT and their dynamic regulation in response to housing temperatures provides a new understanding of the mechanisms underlying BAT thermogenesis. The resources presented in this study offer a valuable platform for future investigations of BAT development and thermogenesis.
Collapse
Affiliation(s)
- Farnaz Shamsi
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02115, USA.
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, 10010, USA.
- Department of Cell Biology, Grossman School of Medicine, New York University, New York, NY, 10016, USA.
| | - Rongbin Zheng
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | - Li-Lun Ho
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kaifu Chen
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA.
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02115, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
11
|
Shi Z, Xiong S, Hu R, Wang Z, Park J, Qian Y, Wang J, Bhalla P, Velupally N, Song Q, Song Z, Layden BT, Jiang Y. The Notch-Pdgfrβ axis suppresses brown adipocyte progenitor differentiation in early postnatal mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.24.541839. [PMID: 37293108 PMCID: PMC10245810 DOI: 10.1101/2023.05.24.541839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
De novo brown adipogenesis holds potential in combating the epidemics of obesity and diabetes. However, the identity of brown adipocyte progenitor cells (APCs) and their regulation have not been extensively studied. Here through in vivo lineage tracing, we observed that PDGFRβ+ pericytes give rise to developmental brown adipocytes, but not to those in adult homeostasis. In contrast, TBX18+ pericytes contribute to brown adipogenesis throughout both developmental and adult stages, though in a depot-specific manner. Mechanistically, Notch inhibition in PDGFRβ+ pericytes promotes brown adipogenesis through the downregulation of PDGFRβ. Furthermore, inhibition of Notch signaling in PDGFRβ+ pericytes mitigates HFHS (high-fat, high-sucrose) induced glucose and metabolic impairment in both developmental and adult stages. Collectively, these findings show that the Notch/PDGFRβ axis negatively regulates developmental brown adipogenesis, and its repression promotes brown adipose tissue expansion and improves metabolic health. Highlights PDGFRβ+ pericytes act as an essential developmental brown APC.TBX18+ pericytes contribute to brown adipogenesis in a depot-specific manner.Inhibiting Notch-Pdgfrβ axis promotes brown APC adipogenesis.Enhanced postnatal brown adipogenesis improves metabolic health in adult stage.
Collapse
|
12
|
Nigro P, Vamvini M, Yang J, Caputo T, Ho LL, Carbone NP, Papadopoulos D, Conlin R, He J, Hirshman MF, White JD, Robidoux J, Hickner RC, Nielsen S, Pedersen BK, Kellis M, Middelbeek RJW, Goodyear LJ. Exercise training remodels inguinal white adipose tissue through adaptations in innervation, vascularization, and the extracellular matrix. Cell Rep 2023; 42:112392. [PMID: 37058410 PMCID: PMC10374102 DOI: 10.1016/j.celrep.2023.112392] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 02/13/2023] [Accepted: 03/30/2023] [Indexed: 04/15/2023] Open
Abstract
Inguinal white adipose tissue (iWAT) is essential for the beneficial effects of exercise training on metabolic health. The underlying mechanisms for these effects are not fully understood, and here, we test the hypothesis that exercise training results in a more favorable iWAT structural phenotype. Using biochemical, imaging, and multi-omics analyses, we find that 11 days of wheel running in male mice causes profound iWAT remodeling including decreased extracellular matrix (ECM) deposition and increased vascularization and innervation. We identify adipose stem cells as one of the main contributors to training-induced ECM remodeling, show that the PRDM16 transcriptional complex is necessary for iWAT remodeling and beiging, and discover neuronal growth regulator 1 (NEGR1) as a link between PRDM16 and neuritogenesis. Moreover, we find that training causes a shift from hypertrophic to insulin-sensitive adipocyte subpopulations. Exercise training leads to remarkable adaptations to iWAT structure and cell-type composition that can confer beneficial changes in tissue metabolism.
Collapse
Affiliation(s)
- Pasquale Nigro
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Maria Vamvini
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA; Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jiekun Yang
- Computational Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tiziana Caputo
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA; Computational Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Li-Lun Ho
- Computational Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nicholas P Carbone
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Danae Papadopoulos
- Computational Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Royce Conlin
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Jie He
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Michael F Hirshman
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Joseph D White
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC, USA
| | - Jacques Robidoux
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC, USA
| | - Robert C Hickner
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC, USA; Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL, USA
| | - Søren Nielsen
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Bente K Pedersen
- The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Manolis Kellis
- Computational Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Roeland J W Middelbeek
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA; Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Abstract
Brown adipose tissue (BAT) displays the unique capacity to generate heat through uncoupled oxidative phosphorylation that makes it a very attractive therapeutic target for cardiometabolic diseases. Here, we review BAT cellular metabolism, its regulation by the central nervous and endocrine systems and circulating metabolites, the plausible roles of this tissue in human thermoregulation, energy balance, and cardiometabolic disorders, and the current knowledge on its pharmacological stimulation in humans. The current definition and measurement of BAT in human studies relies almost exclusively on BAT glucose uptake from positron emission tomography with 18F-fluorodeoxiglucose, which can be dissociated from BAT thermogenic activity, as for example in insulin-resistant states. The most important energy substrate for BAT thermogenesis is its intracellular fatty acid content mobilized from sympathetic stimulation of intracellular triglyceride lipolysis. This lipolytic BAT response is intertwined with that of white adipose (WAT) and other metabolic tissues, and cannot be independently stimulated with the drugs tested thus far. BAT is an interesting and biologically plausible target that has yet to be fully and selectively activated to increase the body's thermogenic response and shift energy balance. The field of human BAT research is in need of methods able to directly, specifically, and reliably measure BAT thermogenic capacity while also tracking the related thermogenic responses in WAT and other tissues. Until this is achieved, uncertainty will remain about the role played by this fascinating tissue in human cardiometabolic diseases.
Collapse
Affiliation(s)
- André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | | | - Denis Richard
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Quebec City, Quebec, G1V 4G5, Canada
| |
Collapse
|
14
|
Qian X, Meng X, Zhang S, Zeng W. Neuroimmune regulation of white adipose tissues. FEBS J 2022; 289:7830-7853. [PMID: 34564950 DOI: 10.1111/febs.16213] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/21/2021] [Accepted: 09/24/2021] [Indexed: 01/14/2023]
Abstract
The white adipose tissues (WAT) are located in distinct depots throughout the body. They serve as an energy reserve, providing fatty acids for other tissues via lipolysis when needed, and function as an endocrine organ to regulate systemic metabolism. Their activities are coordinated through intercellular communications among adipocytes and other cell types such as residential and infiltrating immune cells, which are collectively under neuronal control. The adipocytes and immune subtypes including macrophages/monocytes, eosinophils, neutrophils, group 2 innate lymphoid cells (ILC2s), T and B cells, dendritic cells (DCs), and natural killer (NK) cells display cellular and functional diversity in response to the energy states and contribute to metabolic homeostasis and pathological conditions. Accumulating evidence reveals that neuronal innervations control lipid deposition and mobilization via regulating lipolysis, adipocyte size, and cellularity. Vice versa, the neuronal innervations and activity are influenced by cellular factors in the WAT. Though the literature describing adipose tissue cells is too extensive to cover in detail, we strive to highlight a selected list of neuronal and immune components in this review. The cell-to-cell communications and the perspective of neuroimmune regulation are emphasized to enlighten the potential therapeutic opportunities for treating metabolic disorders.
Collapse
Affiliation(s)
- Xinmin Qian
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Xia Meng
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Shan Zhang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Wenwen Zeng
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China.,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| |
Collapse
|
15
|
Li S, Wang Y, Li Z, Long C, Zhou Q, Chen Q. The links between adipose tissue DNA methylation, obesity, and insulin resistance: A protocol for systematic review. Medicine (Baltimore) 2022; 101:e31261. [PMID: 36451420 PMCID: PMC9704914 DOI: 10.1097/md.0000000000031261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND RATIONALE Obesity is a metabolic condition brought on by the interplay of hereditary and environmental factors, making it one of the most common diseases in the world. Insulin resistance (IR) and obesity have a close connection and can both be advantageous. One of the main methods of epigenetic regulation is DNA methylation modification. Studies have demonstrated over the past few years that DNA methylation is crucial to the emergence of obesity and DNA methylation can lead to IR. Adipose tissue participates in the physiopathological processes of obesity and IR and functions as an endocrine organ controlling the body's balanced metabolism, thus, adipose tissue-associated gene DNA methylation affects the development of obesity and IR by influencing the function of adipose tissue. Hence, an explanation of current research on DNA methylation, IR, and obesity, following the most recent developments, exploring changes in DNA methylation in different types of adipose tissue in insulin-resistant patients and obese patients may enable the identification of novel targets in clinical obesity prevention and treatment. METHOD AND ANALYSIS The following electronic bibliographic databases will be searched from inception for peer-reviewed original research published: MEDLINE (through PubMed), Scopus, and EMBASE. Cochrane Library, Cochrane Clinical Trials Registry, the National Institutes for Health Clinical Trials Registry, and the WHO International Clinical Trials Registry Platform from inception to December 31, 2021 will be conducted. Systematic reviews will adhere to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses reporting guidelines. The development of search strategies will make use of medical issue phrases and keywords associated with DNA methylation, Adipose tissue DNA methylation, obesity, and IR. Identified citations will be independently reviewed by two authors to determine eligibility at the title and abstract level, and then at the full text and data extraction phases. Disagreements and conflicts will be resolved through discussion with a third author. Two authors will extract the necessary data from the included studies independently, and The Cochrane Risk of Bias Assessment Tool will be used to assess the bias of randomized controlled studies, and the Newcastle-Ottawa scale for nonrandomized controlled studies. If the interventions and outcomes evaluated are sufficiently homogeneous, results from subgroups of studies will be pooled together in a meta-analysis.
Collapse
Affiliation(s)
- Suwen Li
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yan Wang
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zinan Li
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Cong Long
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qian Zhou
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qiu Chen
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan, China
- * Correspondence: Qiu Chen Department of Endocrinology, Hospital of Chengdu University of Traditional Chines-e Medicine, Chengdu 610072, China ()
| |
Collapse
|
16
|
Choi CHJ, Barr W, Zaman S, Model C, Park A, Koenen M, Lin Z, Szwed SK, Marchildon F, Crane A, Carroll TS, Molina H, Cohen P. LRG1 is an adipokine that promotes insulin sensitivity and suppresses inflammation. eLife 2022; 11:e81559. [PMID: 36346018 PMCID: PMC9674348 DOI: 10.7554/elife.81559] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 11/06/2022] [Indexed: 11/11/2022] Open
Abstract
While dysregulation of adipocyte endocrine function plays a central role in obesity and its complications, the vast majority of adipokines remain uncharacterized. We employed bio-orthogonal non-canonical amino acid tagging (BONCAT) and mass spectrometry to comprehensively characterize the secretome of murine visceral and subcutaneous white and interscapular brown adip ocytes. Over 600 proteins were identified, the majority of which showed cell type-specific enrichment. We here describe a metabolic role for leucine-rich α-2 glycoprotein 1 (LRG1) as an obesity-regulated adipokine secreted by mature adipocytes. LRG1 overexpression significantly improved glucose homeostasis in diet-induced and genetically obese mice. This was associated with markedly reduced white adipose tissue macrophage accumulation and systemic inflammation. Mechanistically, we found LRG1 binds cytochrome c in circulation to dampen its pro-inflammatory effect. These data support a new role for LRG1 as an insulin sensitizer with therapeutic potential given its immunomodulatory function at the nexus of obesity, inflammation, and associated pathology.
Collapse
Affiliation(s)
- Chan Hee J Choi
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD ProgramNew YorkUnited States
- Laboratory of Molecular Metabolism, Rockefeller UniversityNew YorkUnited States
| | - William Barr
- Laboratory of Molecular Metabolism, Rockefeller UniversityNew YorkUnited States
| | - Samir Zaman
- Laboratory of Molecular Metabolism, Rockefeller UniversityNew YorkUnited States
| | - Corey Model
- Laboratory of Molecular Metabolism, Rockefeller UniversityNew YorkUnited States
| | - Annsea Park
- Department of Immunobiology, Yale UniversityNew HavenUnited States
| | - Mascha Koenen
- Laboratory of Molecular Metabolism, Rockefeller UniversityNew YorkUnited States
| | - Zeran Lin
- Laboratory of Molecular Metabolism, Rockefeller UniversityNew YorkUnited States
| | - Sarah K Szwed
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD ProgramNew YorkUnited States
- Laboratory of Molecular Metabolism, Rockefeller UniversityNew YorkUnited States
| | - Francois Marchildon
- Laboratory of Molecular Metabolism, Rockefeller UniversityNew YorkUnited States
| | - Audrey Crane
- Laboratory of Molecular Metabolism, Rockefeller UniversityNew YorkUnited States
| | - Thomas S Carroll
- Bioinformatics Resouce Center, Rockefeller UniversityNew YorkUnited States
| | - Henrik Molina
- Proteomics Resource Center, Rockefeller UniversityNew YorkUnited States
| | - Paul Cohen
- Laboratory of Molecular Metabolism, Rockefeller UniversityNew YorkUnited States
| |
Collapse
|
17
|
Martinez-Sanchez N, Sweeney O, Sidarta-Oliveira D, Caron A, Stanley SA, Domingos AI. The sympathetic nervous system in the 21st century: Neuroimmune interactions in metabolic homeostasis and obesity. Neuron 2022; 110:3597-3626. [PMID: 36327900 PMCID: PMC9986959 DOI: 10.1016/j.neuron.2022.10.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 08/23/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
The sympathetic nervous system maintains metabolic homeostasis by orchestrating the activity of organs such as the pancreas, liver, and white and brown adipose tissues. From the first renderings by Thomas Willis to contemporary techniques for visualization, tracing, and functional probing of axonal arborizations within organs, our understanding of the sympathetic nervous system has started to grow beyond classical models. In the present review, we outline the evolution of these findings and provide updated neuroanatomical maps of sympathetic innervation. We offer an autonomic framework for the neuroendocrine loop of leptin action, and we discuss the role of immune cells in regulating sympathetic terminals and metabolism. We highlight potential anti-obesity therapeutic approaches that emerge from the modern appreciation of SNS as a neural network vis a vis the historical fear of sympathomimetic pharmacology, while shifting focus from post- to pre-synaptic targeting. Finally, we critically appraise the field and where it needs to go.
Collapse
Affiliation(s)
| | - Owen Sweeney
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Davi Sidarta-Oliveira
- Physician-Scientist Graduate Program, Obesity and Comorbidities Research Center, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Alexandre Caron
- Faculty of Pharmacy, Université Laval, Québec City, QC G1V 0A6, Canada
| | - Sarah A Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ana I Domingos
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK.
| |
Collapse
|
18
|
Chumasov EI, Petrova ES, Korzhevskii DE. Morphological Peculiarities of Innervation of Rat Epicardial Adipose Tissue in Early Postnatal Ontogenesis. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022060333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
19
|
Solivan-Rivera J, Yang Loureiro Z, DeSouza T, Desai A, Pallat S, Yang Q, Rojas-Rodriguez R, Ziegler R, Skritakis P, Joyce S, Zhong D, Nguyen T, Corvera S. A neurogenic signature involving monoamine Oxidase-A controls human thermogenic adipose tissue development. eLife 2022; 11:e78945. [PMID: 36107478 PMCID: PMC9519151 DOI: 10.7554/elife.78945] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 09/09/2022] [Indexed: 11/13/2022] Open
Abstract
Mechanisms that control 'beige/brite' thermogenic adipose tissue development may be harnessed to improve human metabolic health. To define these mechanisms, we developed a species-hybrid model in which human mesenchymal progenitor cells were used to develop white or thermogenic/beige adipose tissue in mice. The hybrid adipose tissue developed distinctive features of human adipose tissue, such as larger adipocyte size, despite its neurovascular architecture being entirely of murine origin. Thermogenic adipose tissue recruited a denser, qualitatively distinct vascular network, differing in genes mapping to circadian rhythm pathways, and denser sympathetic innervation. The enhanced thermogenic neurovascular network was associated with human adipocyte expression of THBS4, TNC, NTRK3, and SPARCL1, which enhance neurogenesis, and decreased expression of MAOA and ACHE, which control neurotransmitter tone. Systemic inhibition of MAOA, which is present in human but absent in mouse adipocytes, induced browning of human but not mouse adipose tissue, revealing the physiological relevance of this pathway. Our results reveal species-specific cell type dependencies controlling the development of thermogenic adipose tissue and point to human adipocyte MAOA as a potential target for metabolic disease therapy.
Collapse
Affiliation(s)
- Javier Solivan-Rivera
- Morningside Graduate School of Biomedical Sciences, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Zinger Yang Loureiro
- Morningside Graduate School of Biomedical Sciences, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Tiffany DeSouza
- Program in Molecular Medicine, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Anand Desai
- Program in Molecular Medicine, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Sabine Pallat
- Morningside Graduate School of Biomedical Sciences, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Qin Yang
- Morningside Graduate School of Biomedical Sciences, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Raziel Rojas-Rodriguez
- Program in Molecular Medicine, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Rachel Ziegler
- Program in Molecular Medicine, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Pantos Skritakis
- Program in Molecular Medicine, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Shannon Joyce
- Program in Molecular Medicine, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Denise Zhong
- Program in Molecular Medicine, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Tammy Nguyen
- Department of Surgery, University of Massachusetts Medical SchoolWorcesterUnited States
- Diabetes Center of Excellence, University of Massachusetts Medical CenterWorcesterUnited States
| | - Silvia Corvera
- Program in Molecular Medicine, University of Massachusetts Medical SchoolWorcesterUnited States
- Diabetes Center of Excellence, University of Massachusetts Medical CenterWorcesterUnited States
| |
Collapse
|
20
|
Chumasov EI, Petrova ES, Korzhevskii DE. Peculiarities of the Innervation of Epicardial Adipose Tissue in a Rat with Aging (Immunohistochemical Study). ADVANCES IN GERONTOLOGY 2022. [DOI: 10.1134/s2079057022030055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
21
|
Fede C, Petrelli L, Pirri C, Neuhuber W, Tiengo C, Biz C, De Caro R, Schleip R, Stecco C. Innervation of human superficial fascia. Front Neuroanat 2022; 16:981426. [PMID: 36106154 PMCID: PMC9464976 DOI: 10.3389/fnana.2022.981426] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
The superficial fascia has only recently been recognized as a specific anatomical structure. Furthermore, whereas it is actually recognized that the innervation of the deep/muscular fascia plays a key role in proprioception and nociception, there are very few studies that have analyzed these characteristics in the superficial fascia. In this work, our group analyzed two different anatomical districts (abdomen and thigh), from volunteer patients, undergoing surgery procedures. Each sample was processed for histological analysis by Hematoxylin&Eosin, and by immunohistochemistry stainings (in 5-micron-paraffin embedded section and in cryosectioned free floating samples), with antibodies specific for nerve fibers: S100 antibody for myelinating and non-myelinating Schwann cells, PGP9.5 antibody as pan-neuronal marker, tyrosine hydroxylase for autonomic innervation. The results revealed a huge innervation: the nervous structures were found above all around blood vessels and close to adipocytes, but they penetrated also in the connective tissue itself and are found in the midst of fibro-adipose tissue. The tissue is pervaded by both thin (mean diameter of 4.8 ± 2.6 μm) and large nerve fiber bundles of greater diameter (21.1 ± 12.2 μm). The ratio S100/TH positivity was equal to 2.96, with a relative percentage of autonomic innervation with of 33.82%. In the light of these findings is evident that the superficial fasciae have a clear and distinct anatomical identity and a specific innervation, which should be considered to better understand their role in thermoregulation, exteroception and pain perception. The knowledge of the superficial fascia may improve grading and developing of different manual approach for treatments of fascial dysfunctions, and the understanding of how some factors like temperature or manual therapies can have an impact on sensitivity of the fascia.
Collapse
Affiliation(s)
- Caterina Fede
- Department of Neurosciences, Institute of Human Anatomy, University of Padua, Padua, Italy
- *Correspondence: Caterina Fede
| | - Lucia Petrelli
- Department of Neurosciences, Institute of Human Anatomy, University of Padua, Padua, Italy
| | - Carmelo Pirri
- Department of Neurosciences, Institute of Human Anatomy, University of Padua, Padua, Italy
| | - Winfried Neuhuber
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Cesare Tiengo
- Plastic and Reconstructive Surgery Unit University of Padua, Padua, Italy
| | - Carlo Biz
- Orthopedics and Orthopedic Oncology, Department of Surgery, Oncology and Gastroenterology DiSCOG, University of Padua, Padua, Italy
| | - Raffaele De Caro
- Department of Neurosciences, Institute of Human Anatomy, University of Padua, Padua, Italy
| | - Robert Schleip
- Department of Sport and Health Sciences, Technical University of Munich, Munich, Germany
| | - Carla Stecco
- Department of Neurosciences, Institute of Human Anatomy, University of Padua, Padua, Italy
| |
Collapse
|
22
|
Meng X, Chen J, Zeng W. Stromal cell-derived NGF controls sympathetic innervation in subcutaneous fat. J Lipid Res 2022; 64:100264. [PMID: 35963456 PMCID: PMC9932450 DOI: 10.1016/j.jlr.2022.100264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/06/2022] [Accepted: 08/08/2022] [Indexed: 02/07/2023] Open
Affiliation(s)
- Xia Meng
- Institute for Immunology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, and Center for Life Sciences, Beijing, China
| | - Jian Chen
- Chinese Institute for Brain Research, Beijing, China.
| | - Wenwen Zeng
- Institute for Immunology and Department of Basic Medical Sciences, School of Medicine, Tsinghua University, and Center for Life Sciences, Beijing, China; Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China.
| |
Collapse
|
23
|
Abstract
Neuronal innervation in the adipose tissues plays a crucial role in regulating adipose thermogenic capacity and metabolic homeostasis. The tissue-wide nerves display a large extent of structural plasticity under physiological and pathological conditions that alter the neuronal control of metabolic states. We find here that neuronal plasticity is regulated by immune cells, which constitutes an appealing way to reshape neural-controlled energy balance by targeting immune components. Sympathetic innervation regulates energy balance, and the nerve density in the adipose tissues changes under various metabolic states, resulting in altered neuronal control and conferring resilience to metabolic challenges. However, the impact of the immune milieu on neuronal innervation is not known. Here, we examined the regulatory role on nerve plasticity by eosinophils and found they increased cell abundance in response to cold and produced nerve growth factor (NGF) in the white adipose tissues (WAT). Deletion of Ngf from eosinophils or depletion of eosinophils impairs cold-induced axonal outgrowth and beiging process. The spatial proximity between sympathetic nerves, IL-33–expressing stromal cells, and eosinophils was visualized in both human and mouse adipose tissues. At the cellular level, the sympathetic adrenergic signal induced calcium flux in the stromal cells and subsequent release of IL-33, which drove the up-regulation of IL-5 from group 2 innate lymphoid cells (ILC2s), leading to eosinophil accretion. We propose a feed-forward loop between sympathetic activity and type 2 immunity that coordinately enhances sympathetic innervation and promotes energy expenditure.
Collapse
|
24
|
Lin Z, Chi J, Cohen P. A Clearing Method for Three-Dimensional Imaging of Adipose Tissue. Methods Mol Biol 2022; 2448:73-82. [PMID: 35167090 DOI: 10.1007/978-1-0716-2087-8_4] [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: 06/14/2023]
Abstract
Adipose tissue is not simply a collection of lipid storing fat cells, but is now recognized to be a complex tissue with a central role in whole body energy homeostasis. In order to understand how adipocytes and associated cell types interact in normal physiology and in pathological states like obesity, it is critical to obtain a holistic view of cells and structures in three dimensions. To that end, we have adapted the iDISCO/iDISCO+ tissue clearing protocol to facilitate the delipidation of fat tissues, while still maintaining their architecture. We describe here this method, that we refer to as Adipo-Clear, highlighting key steps in the protocol as well as important technical considerations. This versatile approach can provide entirely new insights into adipose tissue biology in health and disease.
Collapse
Affiliation(s)
- Zeran Lin
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Jingyi Chi
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
25
|
Bruder J, Fromme T. Global Adipose Tissue Remodeling During the First Month of Postnatal Life in Mice. Front Endocrinol (Lausanne) 2022; 13:849877. [PMID: 35250892 PMCID: PMC8892685 DOI: 10.3389/fendo.2022.849877] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 01/21/2022] [Indexed: 12/13/2022] Open
Abstract
During the first month of postnatal life, adipose tissue depots of mice go through a drastic, but transient, remodeling process. Between postnatal days 10 and 20, several white fat depots display a strong and sudden surge in beige adipocyte emergence that reverts until day 30. At the same time, brown fat depots appear to undergo an opposite phenomenon. We comprehensively describe these events, their depot specificity and known environmental and genetic interactions, such as maternal diet, housing temperature and mouse strain. We further discuss potential mechanisms and plausible purposes, including the tempting hypothesis that postnatal transient remodeling creates a lasting adaptive capacity still detectable in adult animals. Finally, we propose postnatal adipose tissue remodeling as a model process to investigate mechanisms of beige adipocyte recruitment advantageous to cold exposure or adrenergic stimulation in its entirely endogenous sequence of events without external manipulation.
Collapse
Affiliation(s)
- Johanna Bruder
- Else Kröner-Fresenius Center for Nutritional Medicine (EKFZ), Technical University of Munich, Freising, Germany
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Tobias Fromme
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- *Correspondence: Tobias Fromme,
| |
Collapse
|