201
|
Naufahu J, Elliott B, Markiv A, Dunning-Foreman P, McGrady M, Howard D, Watt P, Mackenzie RWA. High-Intensity Exercise Decreases IP6K1 Muscle Content and Improves Insulin Sensitivity (SI2*) in Glucose-Intolerant Individuals. J Clin Endocrinol Metab 2018; 103:1479-1490. [PMID: 29300979 DOI: 10.1210/jc.2017-02019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 12/20/2017] [Indexed: 12/13/2022]
Abstract
CONTEXT Insulin resistance (IR) in skeletal muscle contributes to whole body hyperglycemia and the secondary complications associated with type 2 diabetes. Inositol hexakisphosphate kinase-1 (IP6K1) may inhibit insulin-stimulated glucose transport in this tissue type. OBJECTIVE Muscle and plasma IP6K1 were correlated with two-compartment models of glucose control in insulin-resistant hyperinsulinemic individuals. Muscle IP6K1 was also compared after two different exercise trials. DESIGN Nine prediabetic [hemoglobin A1c; 6.1% (0.2%)] patients were recruited to take part in a resting control, a continuous exercise (90% of lactate threshold), and a high-intensity exercise trial (6 30-second sprints). Muscle biopsies were drawn before and after each 60-minute trial. A labeled ([6,62H2]glucose) intravenous glucose tolerance test was performed immediately after the second muscle sample. RESULTS Fasting muscle IP6K1 content did not correlate with insulin sensitivity (SI2*) (P = 0.961). High-intensity exercise reduced IP6K1 muscle protein and messenger RNA expression (P = 0.001). There was no effect on protein IP6K1 content after continuous exercise. Akt308 phosphorylation of was significantly greater after high-intensity exercise. Intermittent exercise reduced hepatic glucose production after the same trial. The same intervention also increased SI2*, and this effect was significantly greater compared with the effect of continuous exercise improvements. Our in vitro experiment demonstrated that the chemical inhibition of IP6K1 increased insulin signaling in C2C12 myotubes. CONCLUSIONS The in vivo and in vitro approaches used in the current study suggest that a decrease in muscle IP6K1 may be linked to whole body increases in SI2*. In addition, high-intensity exercise reduces hepatic glucose production in insulin-resistant individuals.
Collapse
Affiliation(s)
- Jane Naufahu
- Faculty of Science and Technology, Department of Life Sciences, University of Westminster, London, United Kingdom
| | - Bradley Elliott
- Faculty of Science and Technology, Department of Life Sciences, University of Westminster, London, United Kingdom
| | - Anatoliy Markiv
- Biosciences Education, King's College London, London, United Kingdom
| | - Petra Dunning-Foreman
- Faculty of Science and Technology, Department of Life Sciences, University of Westminster, London, United Kingdom
| | - Maggie McGrady
- Faculty of Science and Technology, Department of Life Sciences, University of Westminster, London, United Kingdom
| | - David Howard
- Department of Oncology, Charing Cross Hospital, Imperial NHS Trust Hospitals, London, United Kingdom
| | - Peter Watt
- Centre for Sport and Exercise Science and Medicine (SESAME), University of Brighton, Eastbourne, United Kingdom
| | | |
Collapse
|
202
|
Ludwig RG, Rocha AL, Mori MA. Circulating molecules that control brown/beige adipocyte differentiation and thermogenic capacity. Cell Biol Int 2018; 42:701-710. [PMID: 29384242 DOI: 10.1002/cbin.10946] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/27/2018] [Indexed: 12/18/2022]
Abstract
Obesity may be counteracted by increased energy expenditure. Circulating molecules act in the adipose tissue to influence brown and beige adipocyte function, differentiation, and thermogenic capacity, which in turn affects substrate utilization and impacts energy balance at the organismal level. These molecules have been envisioned as biomarkers and potential candidates for pharmacological interventions to treat obesity. Here we summarize studies that demonstrate the roles of endogenous circulating molecules of a wide variety in regulating the thermogenic potential of brown and beige fat cells. This review describes the state-of-the-art in the field and helps researchers to prioritize their targets in future studies.
Collapse
Affiliation(s)
- Raissa G Ludwig
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Andréa L Rocha
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Marcelo A Mori
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| |
Collapse
|
203
|
Xu L, Ota T. Emerging roles of SGLT2 inhibitors in obesity and insulin resistance: Focus on fat browning and macrophage polarization. Adipocyte 2018; 7:121-128. [PMID: 29376471 DOI: 10.1080/21623945.2017.1413516] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Obesity-associated low-grade inflammation underlies insulin resistance and associated metabolic comorbidities, such as type 2 diabetes (T2D) and nonalcoholic fatty liver disease. Excessive ectopic fat deposition in obesity causes disorders of energy homeostasis and low-grade chronic inflammation in metabolic tissues. In particular, obesity-induced recruitment and activation of adipose tissue macrophages play a key role in the pathogenesis of insulin resistance and T2D. Therefore, treatment options for energy metabolism and macrophage polarization in obese subjects are needed. Sodium-glucose cotransporter (SGLT) 2 inhibitors increase urinary glucose excretion by inhibiting renal glucose reabsorption, thereby having subsequent anti-hyperglycemic effects and reducing body weight. We recently reported that the SGLT2 inhibitor empagliflozin increases fat utilization and browning in white adipose tissue and attenuates obesity-induced inflammation and insulin resistance by activating M2 macrophages. Thus, this review focuses on the beneficial effects of empagliflozin in energy homeostasis and obesity-related inflammation and insulin resistance.
Collapse
Affiliation(s)
- Liang Xu
- Department of Cell Metabolism and Nutrition, Advanced Preventive Medical Sciences Research Center, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Tsuguhito Ota
- Department of Cell Metabolism and Nutrition, Advanced Preventive Medical Sciences Research Center, Kanazawa University, Kanazawa, Ishikawa, Japan
- Division of Metabolism and Biosystemic Science, Department of Medicine, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| |
Collapse
|
204
|
Lee JH, Yeganeh A, Konoeda H, Moon JH, Sung HK. Flow Cytometry and Lineage Tracing Study for Identification of Adipocyte Precursor Cell (APC) Populations. Methods Mol Biol 2018; 1752:111-121. [PMID: 29564767 DOI: 10.1007/978-1-4939-7714-7_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Flow cytometry and fluorescence-activated cell sorting (FACS) techniques have significantly advanced the characterization of adipocyte precursor cell (APC) populations. They allow immunophenotyping, quantification, and isolation of distinct populations, which is critical for understanding adipose tissue development and homeostasis. Here, we describe the identification and purification of adipocyte precursor cells using flow cytometry and FACS, defined by previously established surface marker profiles. In addition, we describe the mouse models and whole adipose tissue visualization techniques that will enable us to characterize the plasticity and the cellular origin of APCs.
Collapse
Affiliation(s)
- Ju Hee Lee
- Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Azadeh Yeganeh
- Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Hisato Konoeda
- Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Joon Ho Moon
- Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Hoon-Ki Sung
- Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
205
|
Li C, Xu MM, Wang K, Adler AJ, Vella AT, Zhou B. Macrophage polarization and meta-inflammation. Transl Res 2018; 191:29-44. [PMID: 29154757 PMCID: PMC5776711 DOI: 10.1016/j.trsl.2017.10.004] [Citation(s) in RCA: 246] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/13/2017] [Accepted: 10/13/2017] [Indexed: 12/14/2022]
Abstract
Chronic overnutrition and obesity induces low-grade inflammation throughout the body. Termed "meta-inflammation," this chronic state of inflammation is mediated by macrophages located within the colon, liver, muscle, and adipose tissue. A sentinel orchestrator of immune activity and homeostasis, macrophages adopt variable states of activation as a function of time and environmental cues. Meta-inflammation phenotypically skews these polarization states and has been linked to numerous metabolic disorders. The past decade has revealed several key regulators of macrophage polarization, including the signal transducer and activator of transcription family, the peroxisome proliferator-activated receptor gamma, the CCAAT-enhancer-binding proteins (C/EBP) family, and the interferon regulatory factors. Recent studies have also suggested that microRNAs and long noncoding RNA influence macrophage polarization. The pathogenic alteration of macrophage polarization in meta-inflammation is regulated by both extracellular and intracellular cues, resulting in distinct secretome profiles. Meta-inflammation-altered macrophage polarization has been linked to insulin insensitivity, atherosclerosis, inflammatory bowel disease, cancer, and autoimmunity. Thus, further mechanistic exploration into the skewing of macrophage polarization promises to have profound impacts on improving global health.
Collapse
Affiliation(s)
- Chuan Li
- Department of Immunology, University of Connecticut, School of Medicine, Farmington, Conn
| | - Maria M Xu
- Department of Immunology, University of Connecticut, School of Medicine, Farmington, Conn
| | - Kepeng Wang
- Department of Immunology, University of Connecticut, School of Medicine, Farmington, Conn
| | - Adam J Adler
- Department of Immunology, University of Connecticut, School of Medicine, Farmington, Conn
| | - Anthony T Vella
- Department of Immunology, University of Connecticut, School of Medicine, Farmington, Conn.
| | - Beiyan Zhou
- Department of Immunology, University of Connecticut, School of Medicine, Farmington, Conn.
| |
Collapse
|
206
|
Villarroya F, Gavaldà-Navarro A, Peyrou M, Villarroya J, Giralt M. The Lives and Times of Brown Adipokines. Trends Endocrinol Metab 2017; 28:855-867. [PMID: 29113711 DOI: 10.1016/j.tem.2017.10.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/18/2017] [Accepted: 10/19/2017] [Indexed: 12/13/2022]
Abstract
Brown adipose tissue (BAT) is responsible for adaptive non-shivering thermogenesis. Moreover, brown fat secretes regulatory factors, so-called brown adipokines, that have autocrine, paracrine, and endocrine actions. Brown adipokines are either polypeptides or nonpeptidic molecules including lipid molecules and microRNAs. The secretory properties of brown fat are essential for tissue remodeling adaptations to thermogenic necessities. The endocrine properties of brown adipokines are thought to contribute to the association between BAT activity and a healthy metabolic profile in relation to glucose and lipid homeostasis. The identification and characterization of brown adipokines may allow the discovery of circulating biomarkers of BAT activity in humans, and will lead to the development of candidate tools for therapeutic interventions in metabolic diseases.
Collapse
Affiliation(s)
- Francesc Villarroya
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red 'Fisiopatologia de la Obesidad y Nutrición', Madrid, Spain; Institut de Recerca Sant Joan de Déu, Barcelona, Catalonia, Spain.
| | - Aleix Gavaldà-Navarro
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red 'Fisiopatologia de la Obesidad y Nutrición', Madrid, Spain; Institut de Recerca Sant Joan de Déu, Barcelona, Catalonia, Spain
| | - Marion Peyrou
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red 'Fisiopatologia de la Obesidad y Nutrición', Madrid, Spain; Institut de Recerca Sant Joan de Déu, Barcelona, Catalonia, Spain
| | - Joan Villarroya
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain; Institut de Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Catalonia, Spain
| | - Marta Giralt
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red 'Fisiopatologia de la Obesidad y Nutrición', Madrid, Spain; Institut de Recerca Sant Joan de Déu, Barcelona, Catalonia, Spain
| |
Collapse
|
207
|
Nicolas S, Cazareth J, Zarif H, Guyon A, Heurteaux C, Chabry J, Petit-Paitel A. Globular Adiponectin Limits Microglia Pro-Inflammatory Phenotype through an AdipoR1/NF-κB Signaling Pathway. Front Cell Neurosci 2017; 11:352. [PMID: 29184485 PMCID: PMC5694456 DOI: 10.3389/fncel.2017.00352] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/24/2017] [Indexed: 12/14/2022] Open
Abstract
We recently reported that increased levels of Adiponectin (ApN) in the brain led to microglia phenotype and activation state regulation, thus reducing both global brain inflammation and depressive-like behaviors in mice. Apart from this, little is known on ApN molecular effects on microglia, although these cells are crucial in both physiological and pathological processes. Here we fill this gap by studying the effects and targets of ApN toward neuroinflammation. Our findings suggest that ApN deficiency in mice leads to a higher sensitivity of mice to neuroinflammation that is due to enhanced microglia responsiveness to a pro-inflammatory challenge. Moreover, we show that globular ApN (gApN) exerts direct in vivo anti-inflammatory actions on microglia by reducing IL-1β, IL-6, and TNFα synthesis. In vitro, gApN anti-inflammatory properties are confirmed in brain-sorted microglia, primary cultured and microglia cell line (BV2), but are not observed on astrocytes. Our results also show that gApN blocks LPS-induced nitrosative and oxidative stress in microglia. Finally, we demonstrate for the first time that these anti-inflammatory and anti-oxidant actions of gApN on microglia are mediated through an AdipoR1/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Sarah Nicolas
- Centre Nationnal de la Recherche Scientifique, UMR7275 Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, Valbonne, France
| | - Julie Cazareth
- Centre Nationnal de la Recherche Scientifique, UMR7275 Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, Valbonne, France
| | - Hadi Zarif
- Centre Nationnal de la Recherche Scientifique, UMR7275 Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, Valbonne, France
| | - Alice Guyon
- Centre Nationnal de la Recherche Scientifique, UMR7275 Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, Valbonne, France
| | - Catherine Heurteaux
- Centre Nationnal de la Recherche Scientifique, UMR7275 Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, Valbonne, France
| | - Joëlle Chabry
- Centre Nationnal de la Recherche Scientifique, UMR7275 Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, Valbonne, France
| | - Agnès Petit-Paitel
- Centre Nationnal de la Recherche Scientifique, UMR7275 Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, Valbonne, France
| |
Collapse
|
208
|
Dong M, Lin J, Lim W, Jin W, Lee HJ. Role of brown adipose tissue in metabolic syndrome, aging, and cancer cachexia. Front Med 2017; 12:130-138. [PMID: 29119382 DOI: 10.1007/s11684-017-0555-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 04/29/2017] [Indexed: 02/07/2023]
Abstract
Brown adipose tissue (BAT) plays a fundamental role in maintaining body temperature by producing heat. BAT that had been know to exist only in mammals and the human neonate has received great attention for the treatment of obesity and diabetes due to its important function in energy metabolism, ever since it is recently reported that human adults have functional BAT. In addition, beige adipocytes, brown adipocytes in white adipose tissue (WAT), have also been shown to take part in whole body metabolism. Multiple lines of evidence demonstrated that transplantation or activation of BAT or/and beige adipocytes reversed obesity and improved insulin sensitivity. Furthermore, many genes involved in BATactivation and/or the recruitment of beige cells have been found, thereby providing new promising strategies for future clinical application of BAT activation to treat obesity and metabolic diseases. This review focuses on recent advances of BAT function in the metabolic aspect and the relationship between BAT and cancer cachexia, a pathological process accompanied with decreased body weight and increased energy expenditure in cancer patients. The underlying possible mechanisms to reduce BAT mass and its activity in the elderly are also discussed.
Collapse
Affiliation(s)
- Meng Dong
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,The University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Jun Lin
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,The University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Wonchung Lim
- Department of Sports Medicine, College of Health Science, Cheongju University, Cheongju, 363-764, Republic of Korea
| | - Wanzhu Jin
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hyuek Jong Lee
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China. .,Center for Vascular Research, Institute for Basic Science (IBS), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
209
|
Wei Q, Lee JH, Wang H, Bongmba OYN, Wu CS, Pradhan G, Sun Z, Chew L, Bajaj M, Chan L, Chapkin RS, Chen MH, Sun Y. Adiponectin is required for maintaining normal body temperature in a cold environment. BMC PHYSIOLOGY 2017; 17:8. [PMID: 29058611 PMCID: PMC5651620 DOI: 10.1186/s12899-017-0034-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 10/12/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Thermogenic impairment promotes obesity and insulin resistance. Adiponectin is an important regulator of energy homeostasis. While many beneficial metabolic effects of adiponectin resemble that of activated thermogenesis, the role of adiponectin in thermogenesis is not clear. In this study, we investigated the role of adiponectin in thermogenesis using adiponectin-null mice (Adipoq -/-). METHODS Body composition was measured using EchoMRI. Metabolic parameters were determined by indirect calorimetry. Insulin sensitivity was evaluated by glucose- and insulin- tolerance tests. Core body temperature was measured by a TH-8 temperature monitoring system. Gene expression was assessed by real-time PCR and protein levels were analyzed by Western blotting and immunohistochemistry. The mitochondrial density of brown adipose tissue was quantified by calculating the ratio of mtDNA:total nuclear DNA. RESULTS Under normal housing temperature of 24 °C and ad libitum feeding condition, the body weight, body composition, and metabolic profile of Adipoq -/- mice were unchanged. Under fasting condition, Adipoq -/- mice exhibited reduced energy expenditure. Conversely, under cold exposure, Adipoq -/- mice exhibited reduced body temperature, and the expression of thermogenic regulatory genes was significantly reduced in brown adipose tissue (BAT) and subcutaneous white adipose tissue (WAT). Moreover, we observed that mitochondrial content was reduced in BAT and subcutaneous WAT, and the expression of mitochondrial fusion genes was decreased in BAT of Adipoq -/- mice, suggesting that adiponectin ablation diminishes mitochondrial biogenesis and altered mitochondrial dynamics. Our study further revealed that adiponectin deletion suppresses adrenergic activation, and down-regulates β3-adrenergic receptor, insulin signaling, and the AMPK-SIRT1 pathway in BAT. CONCLUSIONS Our findings demonstrate that adiponectin is an essential regulator of thermogenesis, and adiponectin is required for maintaining body temperature under cold exposure.
Collapse
Affiliation(s)
- Qiong Wei
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
- Division of Endocrinology, Zhongda hospital, Southeast University, Nanjing, Jiangsu Province, People's Republic of China, 210002
| | - Jong Han Lee
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
- College of Pharmacy, Gachon University, Incheon, 21936, South Korea
| | - Hongying Wang
- Department of Nutrition and Food Science, Texas A&M University, 214D Cater-Mattil; 2253 TAMU, College Station, TX, 77843, USA
- Laboratory of Lipid & Glucose Metabolism, The First Affiliated Hospital of Chongqing Medical University, Chongqing, Sichuan province, People's Republic of China, 400016
| | - Odelia Y N Bongmba
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chia-Shan Wu
- Department of Nutrition and Food Science, Texas A&M University, 214D Cater-Mattil; 2253 TAMU, College Station, TX, 77843, USA
| | - Geetali Pradhan
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Zilin Sun
- Division of Endocrinology, Zhongda hospital, Southeast University, Nanjing, Jiangsu Province, People's Republic of China, 210002
| | - Lindsey Chew
- Institute of Biosciences and Technology, Houston, TX, 77030, USA
| | - Mandeep Bajaj
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Lawrence Chan
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Robert S Chapkin
- Department of Nutrition and Food Science, Texas A&M University, 214D Cater-Mattil; 2253 TAMU, College Station, TX, 77843, USA
| | - Miao-Hsueh Chen
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yuxiang Sun
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Nutrition and Food Science, Texas A&M University, 214D Cater-Mattil; 2253 TAMU, College Station, TX, 77843, USA.
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
210
|
Macrophage VLDLR mediates obesity-induced insulin resistance with adipose tissue inflammation. Nat Commun 2017; 8:1087. [PMID: 29057873 PMCID: PMC5651811 DOI: 10.1038/s41467-017-01232-w] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 08/25/2017] [Indexed: 12/13/2022] Open
Abstract
Obesity is closely associated with increased adipose tissue macrophages (ATMs), which contribute to systemic insulin resistance and altered lipid metabolism by creating a pro-inflammatory environment. Very low-density lipoprotein receptor (VLDLR) is involved in lipoprotein uptake and storage. However, whether lipid uptake via VLDLR in macrophages affects obesity-induced inflammatory responses and insulin resistance is not well understood. Here we show that elevated VLDLR expression in ATMs promotes adipose tissue inflammation and glucose intolerance in obese mice. In macrophages, VLDL treatment upregulates intracellular levels of C16:0 ceramides in a VLDLR-dependent manner, which potentiates pro-inflammatory responses and promotes M1-like macrophage polarization. Adoptive transfer of VLDLR knockout bone marrow to wild-type mice relieves adipose tissue inflammation and improves insulin resistance in diet-induced obese mice. These findings suggest that increased VLDL-VLDLR signaling in ATMs aggravates adipose tissue inflammation and insulin resistance in obesity. VLDLR regulates cellular lipoprotein uptake and storage. Here, the authors show that VLDLR, expressed on adipose tissue macrophages, is upregulated in obesity and promotes adipose tissue inflammation by upregulating ceramide production and facilitating M1-like macrophage polarization.
Collapse
|
211
|
Human ADMC-Derived Adipocyte Thermogenic Capacity Is Regulated by IL-4 Receptor. Stem Cells Int 2017; 2017:2767916. [PMID: 29158739 PMCID: PMC5660824 DOI: 10.1155/2017/2767916] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 08/24/2017] [Indexed: 11/27/2022] Open
Abstract
Type two innate immune system is anti-inflammatory and may play an important role as the means whereby “browning” is induced in subcutaneous adipocytes. It was shown that IL-4 may influence the fate of adipose cell precursors by promoting differentiation towards more thermogenic adipocytes in mice. Here, we investigated the influence of IL-4 and IL-4 receptor, a type two immune cytokine pathway, on the metabolic activity and thermogenic potential of human adipocytes differentiated from adipose-derived mesenchymal stem cells (ADMSCs) obtained from subcutaneous samples of healthy women undergoing abdominoplasty. Western blot analysis, qPCR, and biochemical analyses were performed 10 days after ADMSC differentiation into mature adipocytes was induced. IL-4 receptor was expressed in both precursor and differentiated adipocytes, and IL-4 treatment increased phosphorylation Y641 of signal transducer and activator of transcription 6 (STAT6) in both cell types. IL-4 treatment also increased expression of thermogenic proteins PGC-1α, UCP-1, and CITED1. In addition, IL-4 increased the secretion of adiponectin, leptin, and FGF21 and promoted lipolysis in differentiated adipocytes. In conclusion, IL-4 may directly modulate differentiation of human adipocytes towards a beige phenotype acting through IL-4 receptors on both adipose precursors and differentiated human adipocytes, metabolic effect that must be considered in some antiallergic drugs.
Collapse
|
212
|
Kim E, Lim SM, Kim MS, Yoo SH, Kim Y. Phyllodulcin, a Natural Sweetener, Regulates Obesity-Related Metabolic Changes and Fat Browning-Related Genes of Subcutaneous White Adipose Tissue in High-Fat Diet-Induced Obese Mice. Nutrients 2017; 9:nu9101049. [PMID: 28934139 PMCID: PMC5691666 DOI: 10.3390/nu9101049] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/25/2017] [Accepted: 09/18/2017] [Indexed: 11/21/2022] Open
Abstract
Phyllodulcin is a natural sweetener found in Hydrangea macrophylla var. thunbergii. This study investigated whether phyllodulcin could improve metabolic abnormalities in high-fat diet (HFD)-induced obese mice. Animals were fed a 60% HFD for 6 weeks to induce obesity, followed by 7 weeks of supplementation with phyllodulcin (20 or 40 mg/kg body weight (b.w.)/day). Stevioside (40 mg/kg b.w./day) was used as a positive control. Phyllodulcin supplementation reduced subcutaneous fat mass, levels of plasma lipids, triglycerides, total cholesterol, and low-density lipoprotein cholesterol and improved the levels of leptin, adiponectin, and fasting blood glucose. In subcutaneous fat tissues, supplementation with stevioside or phyllodulcin significantly decreased mRNA expression of lipogenesis-related genes, including CCAAT/enhancer-binding protein α (C/EBPα), peroxisome proliferator activated receptor γ (PPARγ), and sterol regulatory element-binding protein-1C (SREBP-1c) compared to the high-fat group. Phyllodulcin supplementation significantly increased the expression of fat browning-related genes, including PR domain containing 16 (Prdm16), uncoupling protein 1 (UCP1), and peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α), compared to the high-fat group. Hypothalamic brain-derived neurotrophic factor-tropomyosin receptor kinase B (BDNF-TrkB) signaling was upregulated by phyllodulcin supplementation. In conclusion, phyllodulcin is a potential sweetener that could be used to combat obesity by regulating levels of leptin, fat browning-related genes, and hypothalamic BDNF-TrkB signaling.
Collapse
Affiliation(s)
- Eunju Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea.
| | - Soo-Min Lim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea.
| | - Min-Soo Kim
- Department of Food Science and Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul 05006, Korea.
| | - Sang-Ho Yoo
- Department of Food Science and Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul 05006, Korea.
| | - Yuri Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea.
| |
Collapse
|
213
|
Abstract
Our understanding of adipose tissue as an endocrine organ has been transformed over the last 20 years. During this time, a number of adipocyte-derived factors or adipokines have been identified. This article will review evidence for how adipokines acting via the central nervous system (CNS) regulate normal physiology and disease pathology. The reported CNS-mediated effects of adipokines are varied and include the regulation of energy homeostasis, autonomic nervous system activity, the reproductive axis, neurodevelopment, cardiovascular function, and cognition. Due to the wealth of information available and the diversity of their known functions, the archetypal adipokines leptin and adiponectin will be focused on extensively. Other adipokines with established CNS actions will also be discussed. Due to the difficulties associated with studying CNS function on a molecular level in humans, the majority of our knowledge, and as such the studies described in this paper, comes from work in experimental animal models; however, where possible the relevant data from human studies are also highlighted. © 2017 American Physiological Society. Compr Physiol 7:1359-1406, 2017.
Collapse
Affiliation(s)
- Craig Beall
- Biomedical Neuroscience Research Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, Devon, United Kingdom
| | - Lydia Hanna
- Biomedical Neuroscience Research Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, Devon, United Kingdom
| | - Kate L J Ellacott
- Biomedical Neuroscience Research Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, Devon, United Kingdom
| |
Collapse
|
214
|
Huang Z, Zhong L, Lee JTH, Zhang J, Wu D, Geng L, Wang Y, Wong CM, Xu A. The FGF21-CCL11 Axis Mediates Beiging of White Adipose Tissues by Coupling Sympathetic Nervous System to Type 2 Immunity. Cell Metab 2017; 26:493-508.e4. [PMID: 28844880 DOI: 10.1016/j.cmet.2017.08.003] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 05/24/2017] [Accepted: 08/01/2017] [Indexed: 02/08/2023]
Abstract
Type 2 cytokines are important signals triggering biogenesis of thermogenic beige adipocytes in white adipose tissue (WAT) during cold acclimation. However, how cold activates type 2 immunity in WAT remains obscure. Here we show that cold-induced type 2 immune responses and beiging in subcutaneous WAT (scWAT) are abrogated in mice with adipose-selective ablation of FGF21 or its co-receptor β-Klotho, whereas such impairments are reversed by replenishment with chemokine CCL11. Mechanistically, FGF21 acts on adipocytes in an autocrine manner to promote the expression and secretion of CCL11 via activation of ERK1/2, which drives recruitment of eosinophils into scWAT, leading to increases in accumulation of M2 macrophages, and proliferation and commitment of adipocyte precursors into beige adipocytes. These FGF21-elicited type 2 immune responses and beiging are blocked by CCL11 neutralization. Thus, the adipose-derived FGF21-CCL11 axis triggers cold-induced beiging and thermogenesis by coupling sympathetic nervous system to activation of type 2 immunity in scWAT.
Collapse
Affiliation(s)
- Zhe Huang
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ling Zhong
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jimmy Tsz Hang Lee
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jialiang Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Donghai Wu
- The Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Leiluo Geng
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Chi-Ming Wong
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China.
| | - Aimin Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
215
|
Adipose angiotensin II type 1 receptor-associated protein ameliorates metabolic disorders via promoting adipose tissue adipogenesis and browning. Eur J Cell Biol 2017; 96:567-578. [DOI: 10.1016/j.ejcb.2017.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/12/2017] [Accepted: 05/15/2017] [Indexed: 11/24/2022] Open
|
216
|
Castro É, Silva TEO, Festuccia WT. Critical review of beige adipocyte thermogenic activation and contribution to whole-body energy expenditure. Horm Mol Biol Clin Investig 2017; 31:/j/hmbci.ahead-of-print/hmbci-2017-0042/hmbci-2017-0042.xml. [PMID: 28862985 DOI: 10.1515/hmbci-2017-0042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 07/17/2017] [Indexed: 12/30/2022]
Abstract
Beige (or brite, "brown in white") adipocytes are uncoupling protein 1 (UCP1)-positive cells residing in white adipose depots that, depending on the conditions, behave either as classic white adipocytes, storing energy as lipids, or as brown adipocytes, dissipating energy from oxidative metabolism as heat through non-shivering thermogenesis. Because of their thermogenic potential and, therefore, possible usage to treat metabolic diseases such as obesity and type 2 diabetes, beige cells have attracted the attention of many scientists worldwide aiming to develop strategies to safely recruit and activate their thermogenic activity. Indeed, in recent years, a large variety of conditions, molecules (including nutrients) and signaling pathways were reported to promote the recruitment of beige adipocytes. Despite of those advances, the true contribution of beige adipocyte thermogenesis to whole-body energy expenditure is still not completely defined. Herein, we discuss some important aspects that should be considered when studying beige adipocyte biology and the contribution to energy balance and whole-body metabolism.
Collapse
|
217
|
Nawaz A, Aminuddin A, Kado T, Takikawa A, Yamamoto S, Tsuneyama K, Igarashi Y, Ikutani M, Nishida Y, Nagai Y, Takatsu K, Imura J, Sasahara M, Okazaki Y, Ueki K, Okamura T, Tokuyama K, Ando A, Matsumoto M, Mori H, Nakagawa T, Kobayashi N, Saeki K, Usui I, Fujisaka S, Tobe K. CD206 + M2-like macrophages regulate systemic glucose metabolism by inhibiting proliferation of adipocyte progenitors. Nat Commun 2017; 8:286. [PMID: 28819169 PMCID: PMC5561263 DOI: 10.1038/s41467-017-00231-1] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 06/08/2017] [Indexed: 01/06/2023] Open
Abstract
Adipose tissue resident macrophages have important roles in the maintenance of tissue homeostasis and regulate insulin sensitivity for example by secreting pro-inflammatory or anti-inflammatory cytokines. Here, we show that M2-like macrophages in adipose tissue regulate systemic glucose homeostasis by inhibiting adipocyte progenitor proliferation via the CD206/TGFβ signaling pathway. We show that adipose tissue CD206+ cells are primarily M2-like macrophages, and ablation of CD206+ M2-like macrophages improves systemic insulin sensitivity, which was associated with an increased number of smaller adipocytes. Mice genetically engineered to have reduced numbers of CD206+ M2-like macrophages show a down-regulation of TGFβ signaling in adipose tissue, together with up-regulated proliferation and differentiation of adipocyte progenitors. Our findings indicate that CD206+ M2-like macrophages in adipose tissues create a microenvironment that inhibits growth and differentiation of adipocyte progenitors and, thereby, control adiposity and systemic insulin sensitivity.Adipose tissue contains macrophages that can influence both local and systemic metabolism via the secretion of cytokines. Here, Nawaz et al. report that M2-like macrophages, present in adipose tissue, create a microenvironment that inhibits proliferation of adipocyte progenitors due to the secretion of TGF-β1.
Collapse
Affiliation(s)
- Allah Nawaz
- First Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Aminuddin Aminuddin
- First Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan.,Department of Nutrition, Faculty of Medicine, University of Hasanuddin, Makassar, Kota Makassar, Sulawesi Selatan, 90245, Indonesia
| | - Tomonobu Kado
- First Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Akiko Takikawa
- First Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Seiji Yamamoto
- Department of Pathology, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Koichi Tsuneyama
- Department of Diagnostic Pathology, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan.,Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
| | - Yoshiko Igarashi
- Division of Kampo Diagnostics, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Masashi Ikutani
- Department of Immune Regulation, Research Center for Hepatitis and Immunology, Research Institute, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba, 272-8516, Japan
| | - Yasuhiro Nishida
- First Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Yoshinori Nagai
- Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan.,JST, PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan
| | - Kiyoshi Takatsu
- Department of Immunobiology and Pharmacological Genetics, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan.,Toyama Prefectural Institute for Pharmaceutical Research, 17-1 Nakataikouyama, Imiz-shi, Toyama, 939-0363, Japan
| | - Johji Imura
- Department of Diagnostic Pathology, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Masakiyo Sasahara
- Department of Pathology, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Yukiko Okazaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| | - Kohjiro Ueki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.,Department of Molecular Diabetic Medicine, Diabetes Research Center, National Center for Global Health and Medicine, 1-21-1 Toyama Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan.,Section of Animal Models, Department of Infectious Diseases, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Kumpei Tokuyama
- Doctoral Program in Sports Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8574, Japan
| | - Akira Ando
- Doctoral Program in Sports Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8574, Japan
| | - Michihiro Matsumoto
- Department of Molecular Metabolic Regulation, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Hisashi Mori
- Department of Molecular Neuroscience, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Takashi Nakagawa
- Department of Metabolism and Nutrition, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Norihiko Kobayashi
- Department of Disease Control, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Kumiko Saeki
- Department of Disease Control, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Isao Usui
- First Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan
| | - Shiho Fujisaka
- First Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan.
| | - Kazuyuki Tobe
- First Department of Internal Medicine, University of Toyama, 2630 Sugitani, Toyama-shi, Toyama, 930-0194, Japan.
| |
Collapse
|
218
|
Heil LBB, Silva PL, Pelosi P, Rocco PRM. Immunomodulatory effects of anesthetics in obese patients. World J Crit Care Med 2017; 6:140-152. [PMID: 28828299 PMCID: PMC5547428 DOI: 10.5492/wjccm.v6.i3.140] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 04/27/2017] [Accepted: 07/10/2017] [Indexed: 02/06/2023] Open
Abstract
Anesthesia and surgery have an impact on inflammatory responses, which influences perioperative homeostasis. Inhalational and intravenous anesthesia can alter immune-system homeostasis through multiple processes that include activation of immune cells (such as monocytes, neutrophils, and specific tissue macrophages) with release of pro- or anti-inflammatory interleukins, upregulation of cell adhesion molecules, and overproduction of oxidative radicals. The response depends on the timing of anesthesia, anesthetic agents used, and mechanisms involved in the development of inflammation or immunosuppression. Obese patients are at increased risk for chronic diseases and may have the metabolic syndrome, which features insulin resistance and chronic low-grade inflammation. Evidence has shown that obesity has adverse impacts on surgical outcome, and that immune cells play an important role in this process. Understanding the effects of anesthetics on immune-system cells in obese patients is important to support proper selection of anesthetic agents, which may affect postoperative outcomes. This review article aims to integrate current knowledge regarding the effects of commonly used anesthetic agents on the lungs and immune response with the underlying immunology of obesity. Additionally, it identifies knowledge gaps for future research to guide optimal selection of anesthetic agents for obese patients from an immunomodulatory standpoint.
Collapse
|
219
|
Caputo T, Gilardi F, Desvergne B. From chronic overnutrition to metaflammation and insulin resistance: adipose tissue and liver contributions. FEBS Lett 2017; 591:3061-3088. [DOI: 10.1002/1873-3468.12742] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 06/28/2017] [Accepted: 07/02/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Tiziana Caputo
- Center for Integrative Genomics; Lausanne Faculty of Biology and Medicine; University of Lausanne; Switzerland
| | - Federica Gilardi
- Center for Integrative Genomics; Lausanne Faculty of Biology and Medicine; University of Lausanne; Switzerland
| | - Béatrice Desvergne
- Center for Integrative Genomics; Lausanne Faculty of Biology and Medicine; University of Lausanne; Switzerland
| |
Collapse
|
220
|
de Candia P, De Rosa V, Gigantino V, Botti G, Ceriello A, Matarese G. Immunometabolism of human autoimmune diseases: from metabolites to extracellular vesicles. FEBS Lett 2017. [PMID: 28649760 DOI: 10.1002/1873-3468.12733] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Immunometabolism focuses on the mechanisms regulating the impact of metabolism on lymphocyte activity and autoimmunity outbreak. The adipose tissue is long known to release adipokines, either pro- or anti-inflammatory factors bridging nutrition and immune function. More recently, adipocytes were discovered to also release extracellular vesicles (EVs) containing a plethora of biological molecules, including metabolites and microRNAs, which can regulate cell function/metabolism in distant tissues, suggesting that immune regulatory function by the adipose tissue may be far more complex than originally thought. Moreover, EVs were also identified as important mediators of immune cell-to-cell communication, adding a further microenvironmental mechanism of plasticity to fine-tune specific lymphocyte responses. This Review will first focus on the known mechanisms by which metabolism impacts immune function, presenting a systemic (nutrition and long-ranged adipokines) and a cellular point of view (metabolic pathway derangement in autoimmunity). It will then discuss the new discoveries concerning how EVs may act as nanometric vehicles integrating immune/metabolic responses at the level of the extracellular environment and affecting pathological processes.
Collapse
Affiliation(s)
| | - Veronica De Rosa
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Naples, Italy
| | | | - Gerardo Botti
- IRCCS Istituto Nazionale Tumori, Fondazione G. Pascale, Naples, Italy
| | | | - Giuseppe Matarese
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), Naples, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli 'Federico II', Naples, Italy
| |
Collapse
|
221
|
Abstract
Interactions between macrophages and adipocytes influence both metabolism and inflammation. Obesity-induced changes to macrophages and adipocytes lead to chronic inflammation and insulin resistance. This paper reviews the various functions of macrophages in lean and obese adipose tissue and how obesity alters adipose tissue macrophage phenotypes. Metabolic disease and insulin resistance shift the balance between numerous pro- and anti-inflammatory regulators of macrophages and create a feed-forward loop of increasing inflammatory macrophage activation and worsening adipocyte dysfunction. This ultimately leads to adipose tissue fibrosis and diabetes. The molecular mechanisms underlying these processes have therapeutic implications for obesity, metabolic syndrome, and diabetes.
Collapse
Affiliation(s)
- Dylan Thomas
- Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston Medical Center, 88 East Newton Street, H-3600, Boston, MA 02118.
| | - Caroline Apovian
- Section of Endocrinology, Diabetes, Nutrition and Weight Management, Boston Medical Center, 88 East Newton Street, Robinson 4400, Boston, MA 02118.
| |
Collapse
|
222
|
Than A, Xu S, Li R, Leow MKS, Sun L, Chen P. Angiotensin type 2 receptor activation promotes browning of white adipose tissue and brown adipogenesis. Signal Transduct Target Ther 2017; 2:17022. [PMID: 29263921 PMCID: PMC5661636 DOI: 10.1038/sigtrans.2017.22] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/29/2017] [Accepted: 03/31/2017] [Indexed: 01/06/2023] Open
Abstract
Brown adipose tissue dissipates energy in the form of heat. Recent studies have shown that adult humans possess both classical brown and beige adipocytes (brown-like adipocytes in white adipose tissue, WAT), and stimulating brown and beige adipocyte formation can be a new avenue to treat obesity. Angiotensin II (AngII) is a peptide hormone that plays important roles in energy metabolism via its angiotensin type 1 or type 2 receptors (AT1R and AT2R). Adipose tissue is a major source of AngII and expresses both types of its receptors, implying the autocrine and paracrine role of AngII in regulating adipose functions and self-remodeling. Here, based on the in vitro studies on primary cultures of mouse white adipocytes, we report that, AT2R activation, either by AngII or AT2R agonist (C21), induces white adipocyte browning, by increasing PPARγ expression, at least in part, via ERK1/2, PI3kinase/Akt and AMPK signaling pathways. It is also found that AngII–AT2R enhances brown adipogenesis. In the in vivo studies on mice, administration of AT1R antagonist (ZD7155) or AT2R agonist (C21) leads to the increase of WAT browning, body temperature and serum adiponectin, as well as the decrease of WAT mass and the serum levels of TNFα, triglycerides and free fatty acids. In addition, AT2R-induced browning effect is also observed in human white adipocytes, as evidenced by the increased UCP1 expression and oxygen consumption. Finally, we provide evidence that AT2R plays important roles in hormone T3-induced white adipose browning. This study, for the first time, reveals the browning and brown adipogenic effects of AT2R and suggests a potential therapeutic target to combat obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Aung Than
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Shaohai Xu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore.,Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Ru Li
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | | | - Lei Sun
- Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Peng Chen
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
223
|
Lin YW, Wei LN. Innate immunity orchestrates adipose tissue homeostasis. Horm Mol Biol Clin Investig 2017; 31:hmbci-2017-0013. [PMID: 28672736 DOI: 10.1515/hmbci-2017-0013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 05/01/2017] [Indexed: 02/07/2023]
Abstract
Obesity is strongly associated with multiple diseases including insulin resistance, type 2 diabetes, cardiovascular diseases, fatty liver disease, neurodegenerative diseases and cancers, etc. Adipose tissue (AT), mainly brown AT (BAT) and white AT (WAT), is an important metabolic and endocrine organ that maintains whole-body homeostasis. BAT contributes to non-shivering thermogenesis in a cold environment; WAT stores energy and produces adipokines that fine-tune metabolic and inflammatory responses. Obesity is often characterized by over-expansion and inflammation of WAT where inflammatory cells/mediators are abundant, especially pro-inflammatory (M1) macrophages, resulting in chronic low-grade inflammation and leading to insulin resistance and metabolic complications. Macrophages constitute the major component of innate immunity and can be activated as a M1 or M2 (anti-inflammatory) phenotype in response to environmental stimuli. Polarized M1 macrophage causes AT inflammation, whereas polarized M2 macrophage promotes WAT remodeling into the BAT phenotype, also known as WAT browning/beiging, which enhances insulin sensitivity and metabolic health. This review will discuss the regulation of AT homeostasis in relation to innate immunity.
Collapse
Affiliation(s)
- Yi-Wei Lin
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Li-Na Wei
- Department of Pharmacology, University of Minnesota Medical School, 612 Jackson Hall, 321 Church St. SE., Minneapolis, MN 55455, USA, Phone: 612-625-9402, Fax: 612-625-8408
| |
Collapse
|
224
|
Yeo CR, Agrawal M, Hoon S, Shabbir A, Shrivastava MK, Huang S, Khoo CM, Chhay V, Yassin MS, Tai ES, Vidal-Puig A, Toh SA. SGBS cells as a model of human adipocyte browning: A comprehensive comparative study with primary human white subcutaneous adipocytes. Sci Rep 2017; 7:4031. [PMID: 28642596 PMCID: PMC5481408 DOI: 10.1038/s41598-017-04369-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 05/19/2017] [Indexed: 01/20/2023] Open
Abstract
The Simpson Golabi Behmel Syndrome (SGBS) pre-adipocyte cell strain is widely considered to be a representative in vitro model of human white pre-adipocytes. A recent study suggested that SGBS adipocytes exhibit an unexpected transient brown phenotype. Here, we comprehensively examined key differences between SGBS adipocytes and primary human white subcutaneous (PHWSC) adipocytes. RNA-Seq analysis revealed that extracellular matrix (ECM)-receptor interaction and metabolic pathways were the top two KEGG pathways significantly enriched in SGBS adipocytes, which included positively enriched mitochondrial respiration and oxidation pathways. Compared to PHWSC adipocytes, SGBS adipocytes showed not only greater induction of adipogenic gene expression during differentiation but also increased levels of UCP1 mRNA and protein expression. Functionally, SGBS adipocytes displayed higher ISO-induced basal leak respiration and overall oxygen consumption rate, along with increased triglyceride accumulation and insulin-stimulated glucose uptake. In conclusion, we confirmed that SGBS adipocytes, which are considered of white adipose tissue origin can shift towards a brown/beige adipocyte phenotype. These differences indicate SGBS cells may help to identify mechanisms leading to browning, and inform our understanding for the use of SGBS vis-à-vis primary human subcutaneous adipocytes as a human white adipocyte model, guiding the selection of appropriate cell models in future metabolic research.
Collapse
Affiliation(s)
- Chia Rou Yeo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore
| | - Madhur Agrawal
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore
| | - Shawn Hoon
- Molecular Engineering Laboratory, Biomedical Sciences Institutes, A*Star, 138668, Singapore, Singapore
| | - Asim Shabbir
- Department of Surgery, National University Hospital, 119074, Singapore, Singapore
| | - Manu Kunaal Shrivastava
- Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Shiqi Huang
- Food Science and Technology Program, Department of Chemistry, National University of Singapore, Singapore, 117542, Singapore
| | - Chin Meng Khoo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore
- Department of Medicine, National University Health System, 119228, Singapore, Singapore
| | - Vanna Chhay
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore
| | - M Shabeer Yassin
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore
| | - E Shyong Tai
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore
- Department of Medicine, National University Health System, 119228, Singapore, Singapore
| | - Antonio Vidal-Puig
- Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Sue-Anne Toh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore, Singapore.
- Department of Medicine, National University Health System, 119228, Singapore, Singapore.
| |
Collapse
|
225
|
Weiner J, Rohde K, Krause K, Zieger K, Klöting N, Kralisch S, Kovacs P, Stumvoll M, Blüher M, Böttcher Y, Heiker JT. Brown adipose tissue (BAT) specific vaspin expression is increased after obesogenic diets and cold exposure and linked to acute changes in DNA-methylation. Mol Metab 2017; 6:482-493. [PMID: 28580279 PMCID: PMC5444018 DOI: 10.1016/j.molmet.2017.03.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/10/2017] [Accepted: 03/15/2017] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Several studies have demonstrated anti-diabetic and anti-obesogenic properties of visceral adipose tissue-derived serine protease inhibitor (vaspin) and so evoked its potential use for treatment of obesity-related diseases. The aim of the study was to unravel physiological regulators of vaspin expression and secretion with a particular focus on its role in brown adipose tissue (BAT) biology. METHODS We analyzed the effects of obesogenic diets and cold exposure on vaspin expression in liver and white and brown adipose tissue (AT) and plasma levels. Vaspin expression was analyzed in isolated white and brown adipocytes during adipogenesis and in response to adrenergic stimuli. DNA-methylation within the vaspin promoter was analyzed to investigate acute epigenetic changes after cold-exposure in BAT. RESULTS Our results demonstrate a strong induction of vaspin mRNA and protein expression specifically in BAT of both cold-exposed and high-fat (HF) or high-sugar (HS) fed mice. While obesogenic diets also upregulated hepatic vaspin mRNA levels, cold exposure tended to increase vaspin gene expression of inguinal white adipose tissue (iWAT) depots. Concomitantly, vaspin plasma levels were decreased upon obesogenic or thermogenic triggers. Vaspin expression was increased during adipogenesis but unaffected by sympathetic activation in brown adipocytes. Analysis of vaspin promoter methylation in AT revealed lowest methylation levels in BAT, which were acutely reduced after cold exposure. CONCLUSIONS Our data demonstrate a novel BAT-specific regulation of vaspin gene expression upon physiological stimuli in vivo with acute epigenetic changes that may contribute to cold-induced expression in BAT. We conclude that these findings indicate functional relevance and potentially beneficial effects of vaspin in BAT function.
Collapse
Affiliation(s)
- Juliane Weiner
- Divisions of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
- Institute of Biochemistry, Faculty of Biosciences, Pharmacy and Psychology, University of Leipzig, Leipzig, Germany
| | - Kerstin Rohde
- Leipzig University Medical Center, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
- Department of Clinical Molecular Biology, University of Oslo, Oslo, Norway
| | - Kerstin Krause
- Divisions of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
| | - Konstanze Zieger
- Institute of Biochemistry, Faculty of Biosciences, Pharmacy and Psychology, University of Leipzig, Leipzig, Germany
| | - Nora Klöting
- Divisions of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
- Leipzig University Medical Center, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Susan Kralisch
- Divisions of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
- Leipzig University Medical Center, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Peter Kovacs
- Leipzig University Medical Center, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Michael Stumvoll
- Divisions of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Divisions of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
- Leipzig University Medical Center, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Yvonne Böttcher
- Leipzig University Medical Center, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
- Department of Clinical Molecular Biology, University of Oslo, Oslo, Norway
| | - John T. Heiker
- Divisions of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
- Institute of Biochemistry, Faculty of Biosciences, Pharmacy and Psychology, University of Leipzig, Leipzig, Germany
- Leipzig University Medical Center, IFB Adiposity Diseases, University of Leipzig, Leipzig, Germany
| |
Collapse
|
226
|
Xu L, Nagata N, Nagashimada M, Zhuge F, Ni Y, Chen G, Mayoux E, Kaneko S, Ota T. SGLT2 Inhibition by Empagliflozin Promotes Fat Utilization and Browning and Attenuates Inflammation and Insulin Resistance by Polarizing M2 Macrophages in Diet-induced Obese Mice. EBioMedicine 2017; 20:137-149. [PMID: 28579299 PMCID: PMC5478253 DOI: 10.1016/j.ebiom.2017.05.028] [Citation(s) in RCA: 361] [Impact Index Per Article: 45.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 05/24/2017] [Accepted: 05/25/2017] [Indexed: 12/20/2022] Open
Abstract
Sodium-glucose cotransporter (SGLT) 2 inhibitors increase urinary glucose excretion (UGE), leading to blood glucose reductions and weight loss. However, the impacts of SGLT2 inhibition on energy homeostasis and obesity-induced insulin resistance are less well known. Here, we show that empagliflozin, a SGLT2 inhibitor, enhanced energy expenditure and attenuated inflammation and insulin resistance in high-fat-diet-induced obese (DIO) mice. C57BL/6J mice were pair-fed a high-fat diet (HFD) or a HFD with empagliflozin for 16weeks. Empagliflozin administration increased UGE in the DIO mice, whereas it suppressed HFD-induced weight gain, insulin resistance, and hepatic steatosis. Moreover, empagliflozin shifted energy metabolism towards fat utilization, elevated AMP-activated protein kinase and acetyl-CoA carbolxylase phosphorylation in skeletal muscle, and increased hepatic and plasma fibroblast growth factor 21 levels. Importantly, empagliflozin increased energy expenditure, heat production, and the expression of uncoupling protein 1 in brown fat and in inguinal and epididymal white adipose tissue (WAT). Furthermore, empagliflozin reduced M1-polarized macrophage accumulation while inducing the anti-inflammatory M2 phenotype of macrophages within WAT and liver, lowering plasma TNFα levels and attenuating obesity-related chronic inflammation. Thus, empagliflozin suppressed weight gain by enhancing fat utilization and browning and attenuated obesity-induced inflammation and insulin resistance by polarizing M2 macrophages in WAT and liver.
Collapse
Affiliation(s)
- Liang Xu
- Department of Cell Metabolism and Nutrition, Brain/Liver Interface Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Naoto Nagata
- Department of Cell Metabolism and Nutrition, Brain/Liver Interface Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Mayumi Nagashimada
- Department of Cell Metabolism and Nutrition, Brain/Liver Interface Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Fen Zhuge
- Department of Cell Metabolism and Nutrition, Brain/Liver Interface Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Yinhua Ni
- Department of Cell Metabolism and Nutrition, Brain/Liver Interface Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Guanliang Chen
- Department of Cell Metabolism and Nutrition, Brain/Liver Interface Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Eric Mayoux
- Boehringer-Ingelheim, Cardio-metabolic Diseases Research, Biberach, Germany
| | - Shuichi Kaneko
- Department of System Biology, Kanazawa University Graduate School of Medical Science, Kanazawa, Ishikawa 920-8640, Japan
| | - Tsuguhito Ota
- Department of Cell Metabolism and Nutrition, Brain/Liver Interface Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan; Department of System Biology, Kanazawa University Graduate School of Medical Science, Kanazawa, Ishikawa 920-8640, Japan.
| |
Collapse
|
227
|
Montanari T, Pošćić N, Colitti M. Factors involved in white-to-brown adipose tissue conversion and in thermogenesis: a review. Obes Rev 2017; 18:495-513. [PMID: 28187240 DOI: 10.1111/obr.12520] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/10/2017] [Accepted: 01/11/2017] [Indexed: 12/21/2022]
Abstract
Obesity is the result of energy intake chronically exceeding energy expenditure. Classical treatments against obesity do not provide a satisfactory long-term outcome for the majority of patients. After the demonstration of functional brown adipose tissue in human adults, great effort is being devoted to develop therapies based on the adipose tissue itself, through the conversion of fat-accumulating white adipose tissue into energy-dissipating brown adipose tissue. Anti-obesity treatments that exploit endogenous, pharmacological and nutritional factors to drive such conversion are especially in demand. In the present review, we summarize the current knowledge about the various molecules that can be applied in promoting white-to-brown adipose tissue conversion and energy expenditure and the cellular mechanisms involved.
Collapse
Affiliation(s)
- T Montanari
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Udine, Italy
| | - N Pošćić
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Udine, Italy
| | - M Colitti
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Udine, Italy
| |
Collapse
|
228
|
Hu T, Yuan X, Ye R, Zhou H, Lin J, Zhang C, Zhang H, Wei G, Dong M, Huang Y, Lim W, Liu Q, Lee HJ, Jin W. Brown adipose tissue activation by rutin ameliorates polycystic ovary syndrome in rat. J Nutr Biochem 2017; 47:21-28. [PMID: 28501702 DOI: 10.1016/j.jnutbio.2017.04.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 04/12/2017] [Accepted: 04/15/2017] [Indexed: 12/27/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a complex endocrinopathy that is characterized by anovulation, hyperandrogenism and polycystic ovary. However, there is a lack of effective treatment for PCOS at present because the pathologic cause of PCOS has not been elucidated. Although it has been known that brown adipose tissue transplantation ameliorates PCOS by activating endogenous BAT, BAT transplantation is not applicable in clinic. Therefore, BAT activation with natural compound could be an effective treatment strategy for PCOS patients. Here, we found that 3 weeks of rutin (a novel compound for BAT activation) treatment increased BAT activation, thereby it improved thermogenesis and systemic insulin sensitivity in dehydroepiandrosterone (DHEA)-induced PCOS rat. In addition, the expression levels of ovarian steroidogenic enzymes such as P450C17, aromatase, 3β-HSD, 17β-HSD and STAR were up-regulated in rutin-treated PCOS rat. Furthermore, acyclicity and the serum level of luteinizing hormone were normalized, and a large number of mature ovulated follicle with a reduction of cystic formation were observed in PCOS rat after rutin treatment. Finally, rutin treatment surprisingly improved fertility and birth defect in PCOS rat. Collectively, our results indicate that rutin treatment significantly improves systemic insulin resistance and ovarian malfunction in PCOS, and our findings in this study provide a novel therapeutic option for the treatment of PCOS by activating BAT with rutin.
Collapse
Affiliation(s)
- Tao Hu
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; The University of the Chinese Academy of Sciences, Beijing, 100049, China; Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Xiaoxue Yuan
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; The University of the Chinese Academy of Sciences, Beijing, 100049, China; Institutes of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, 8 East Jingshun St., Beijing, 100015, China
| | - Rongcai Ye
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; The University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Huiqiao Zhou
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; The University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Jun Lin
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; The University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Chuanhai Zhang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hanlin Zhang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; The University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Gang Wei
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; The University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Meng Dong
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; The University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuanyuan Huang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; The University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Wonchung Lim
- Department of Sports Medicine, College of Health Science, Cheongju University, Cheongju, 363-764, Republic of Korea
| | - Qingsong Liu
- High Magnetic Field Laboratory, Chinese Academy of Sciences, Hefei, Anhui, 230031, China
| | - Hyuek Jong Lee
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Wanzhu Jin
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
229
|
Alternatively activated macrophages do not synthesize catecholamines or contribute to adipose tissue adaptive thermogenesis. Nat Med 2017; 23:623-630. [PMID: 28414329 PMCID: PMC5420449 DOI: 10.1038/nm.4316] [Citation(s) in RCA: 272] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 03/06/2017] [Indexed: 12/12/2022]
Abstract
Adaptive thermogenesis is the process of heat generation in response to
cold stimulation and is under the control of the sympathetic nervous system
whose chief effector is the catecholamine norepinephrine (NE). NE enhances
thermogenesis through beta3 adrenergic receptors to activate brown adipose
tissue and by “browning” white adipose tissue. Recent studies
reported that the alternative activation of macrophages in response to IL-4
stimulation induces the expression of tyrosine hydroxylase (TH), a key enzyme in
the catecholamine synthesis pathway, and to provide an alternative source of
locally produced catecholamines during the thermogenic process. We here report
that the deletion of Th in hematopoetic cells of adult mice
neither alters energy expenditure upon cold exposure nor reduces browning in
inguinal adipose tissue. Bone marrow-derived macrophages did not release NE in
response to stimulation with Interleukin-4 (IL-4), and conditioned media from
IL-4 stimulated macrophages failed to induce expression of thermogenic genes,
such as the one for uncoupling protein 1 (Ucp1) in adipocytes
cultured with the conditioned media. Further, chronic IL-4 treatment failed to
increase energy expenditure in WT, Ucp1-/- and
Il4ra-/- mice. Consistent with these findings,
adipose tissue-resident macrophages did not express TH. Thus, we conclude that
alternatively activated macrophages do not synthesize relevant amounts of
catecholamines and hence are not likely to play a direct role in adipocyte
metabolism or adaptive thermogenesis.
Collapse
|
230
|
Shan B, Wang X, Wu Y, Xu C, Xia Z, Dai J, Shao M, Zhao F, He S, Yang L, Zhang M, Nan F, Li J, Liu J, Liu J, Jia W, Qiu Y, Song B, Han JDJ, Rui L, Duan SZ, Liu Y. The metabolic ER stress sensor IRE1α suppresses alternative activation of macrophages and impairs energy expenditure in obesity. Nat Immunol 2017; 18:519-529. [DOI: 10.1038/ni.3709] [Citation(s) in RCA: 217] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 02/13/2017] [Indexed: 02/07/2023]
|
231
|
Park AY, Cha S. Effects of cold sensitivity in the extremities on circulating adiponectin levels and metabolic syndrome in women. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:150. [PMID: 28279166 PMCID: PMC5345261 DOI: 10.1186/s12906-017-1658-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 03/03/2017] [Indexed: 12/15/2022]
Abstract
Background In adipose tissues, adipokine levels, including adiponectin and leptin, are involved in insulin sensitivity and are reciprocally induced by cold temperature stress. Thermogenic response in the extremities (hands and feet) against cold stress can be negatively related to fat mass accumulation, particularly in the abdomen. However, the relationship between the sensation of cold in the extremities and circulating levels of adipokines is not fully understood. Here, we investigated whether adipokine levels are associated with cold hypersensitivity in the hands and feet (CHHF), independent of body mass, and whether the CHHF is related to metabolic syndrome (MS). Methods Associations of the CHHF with serum levels of adipokines and MS risk were evaluated in 1021 Koreans (372 men and 649 women), using a linear regression model while controlling for thermogenic factors and a logistic regression model, respectively. Results The adiponectin levels were positively associated with the CHHF, particularly in women, irrespective of thermogenic factors, including body mass index (β = 1.23 μg/mL, 95% confidence interval [1.04–1.45]). Logistic regression analysis for MS risk via the CHHF showed that there was a significant inverse association in women (odds ratio = 0.449, 95% confidence interval [0.273–0.737]). Conclusions In summary, our founding indicated that the CHHF could induce increased levels of circulating adiponectin and in turn reduce the MS risk in women. Despite complaints of feeling cold, these women could be at lower risk of cardiovascular disease.
Collapse
|
232
|
Hui X, Zhang M, Gu P, Li K, Gao Y, Wu D, Wang Y, Xu A. Adipocyte SIRT1 controls systemic insulin sensitivity by modulating macrophages in adipose tissue. EMBO Rep 2017; 18:645-657. [PMID: 28270525 DOI: 10.15252/embr.201643184] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 02/05/2017] [Accepted: 02/07/2017] [Indexed: 11/09/2022] Open
Abstract
Adipose tissue inflammation, characterized by augmented infiltration and altered polarization of macrophages, contributes to insulin resistance and its associated metabolic diseases. The NAD+-dependent deacetylase SIRT1 serves as a guardian against metabolic disorders in multiple tissues. To dissect the roles of SIRT1 in adipose tissues, metabolic phenotypes of mice with selective ablation of SIRT1 in adipocytes and myeloid cells were monitored. Compared to myeloid-specific SIRT1 depletion, mice with adipocyte-selective deletion of SIRT1 are more susceptible to diet-induced insulin resistance. The phenotypic changes in adipocyte-selective SIRT1 knockout mice are associated with an increased number of adipose-resident macrophages and their polarization toward the pro-inflammatory M1 subtype. Mechanistically, SIRT1 in adipocytes modulates expression and secretion of several adipokines, including adiponectin, MCP-1, and interleukin 4, which in turn alters recruitment and polarization of the macrophages in adipose tissues. In adipocytes, SIRT1 deacetylates the transcription factor NFATc1 and thereby enhances the binding of NFATc1 to the Il4 gene promoter. These findings suggest that adipocyte SIRT1 controls systemic glucose homeostasis and insulin sensitivity via the cross talk with adipose-resident macrophages.
Collapse
Affiliation(s)
- Xiaoyan Hui
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Mingliang Zhang
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Ping Gu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China.,Department of Endocrinology, School of Medicine, Nanjing University Nanjing General Hospital of Nanjing Military Command, Nanjing, China
| | - Kuai Li
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yuan Gao
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Donghai Wu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China .,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China .,Department of Medicine, The University of Hong Kong, Hong Kong, China.,Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
233
|
Zhang Q, Hao H, Xie Z, Cheng Y, Yin Y, Xie M, Huang H, Gao J, Liu H, Tong C, Zang L, Mu Y, Han W. M2 macrophages infusion ameliorates obesity and insulin resistance by remodeling inflammatory/macrophages' homeostasis in obese mice. Mol Cell Endocrinol 2017; 443:63-71. [PMID: 28069536 DOI: 10.1016/j.mce.2017.01.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 02/04/2023]
Abstract
OBJECTIVE The role of M2 macrophages infusion in dealing with obesity is still little known. In this study, the therapeutic effects of M2 macrophages infusion were investigated. METHODS High fat diet (HFD) was used to induce obesity in C57BL/6N mice. 5 × 105 M2 macrophages, derived from the bone marrow, were injected into obese mice through the tail vein twice with an interval of one week. RESULTS One week after the second injection, weight of inguinal adipose pad was significantly decreased. Accordingly, the adipocyte size of epididymal and inguinal adipose tissue (EAT and INAT) shrank. To our interest, we found that the infused M2 macrophages were homed to EAT, reversing the disturbed homeostasis of high M1 to low M2 in obese mice. Meanwhile, EAT with remodeled macrophages' homeostasis expressed less MCP-1, accompanying with decreased recruitment of inflammatory CCR2+CX3CR1lowLy6C+ monocytes from the blood in M2 infusion group. Further, increased M2 in EAT contribute to enhanced expression of UCP1 expression in EAT, which helped to ameliorate insulin resistance and, subsequently, improve the serum level of triglycerides (TG) and low density lipoprotein cholesterol (LDL-c). CONCLUSIONS These findings highlighted that M2 macrophages infusion could ameliorate obesity as well as obesity-related insulin resistance, suggesting an effective and healthy weight loss strategy.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Endocrinology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China; School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China.
| | - Haojie Hao
- Department of Molecular Biology, Institute of Basic Medicine, College of Life Science, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Zongyan Xie
- China-Japan Friendship Hospital, East Yinghuayuan Street, Beijing, 100029, China
| | - Yu Cheng
- Department of Endocrinology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Yaqi Yin
- Department of Endocrinology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Min Xie
- Department of Endocrinology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Hong Huang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Jieqing Gao
- Department of Endocrinology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Hongyu Liu
- Department of Neurosurgery, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Chuan Tong
- Department of Molecular Biology, Institute of Basic Medicine, College of Life Science, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Li Zang
- Department of Endocrinology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Yiming Mu
- Department of Endocrinology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Weidong Han
- Department of Molecular Biology, Institute of Basic Medicine, College of Life Science, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
234
|
Shin W, Okamatsu-Ogura Y, Machida K, Tsubota A, Nio-Kobayashi J, Kimura K. Impaired adrenergic agonist-dependent beige adipocyte induction in aged mice. Obesity (Silver Spring) 2017; 25:417-423. [PMID: 28026903 DOI: 10.1002/oby.21727] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 10/06/2016] [Accepted: 11/07/2016] [Indexed: 01/08/2023]
Abstract
OBJECTIVE There are two types of thermogenic adipocytes expressing uncoupling protein (UCP)-1: the brown adipocyte activated by adrenergic stimulation and the beige adipocyte that appears within the white adipose tissue (WAT) in response to chronic adrenergic stimulation. This study examined age-related changes in responses of both types of adipocytes to adrenergic stimulation in mice. METHODS Aged (age 20 months) and young (4 months) mice were injected daily with either saline or β3-adrenergic receptor agonist CL316,243 (CL; 0.1 mg/kg, once a day) for 1 week. RESULTS The body and WAT weight tended to be higher in aged mice. CL treatment increased UCP-1 protein amounts in both brown adipose tissue and inguinal WAT, suggesting activation of brown and beige adipocytes. However, induction of beige adipocytes was impaired in aged mice, whereas brown adipocyte activation was comparable to young mice. The number of platelet-derived growth factor receptor α-expressing progenitor cells, which were reported to differentiate into beige adipocytes, significantly decreased in inguinal WAT of aged mice compared with that of young mice. CONCLUSIONS Inductive ability of beige adipocytes in WAT declines with aging in mice. It may be partly because of a decreased number of progenitor cells associated with aging.
Collapse
Affiliation(s)
- Woongchul Shin
- Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yuko Okamatsu-Ogura
- Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Ken Machida
- Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Ayumi Tsubota
- Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Junko Nio-Kobayashi
- Laboratory of Histology and Cytology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kazuhiro Kimura
- Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
235
|
van den Berg SM, van Dam AD, Rensen PCN, de Winther MPJ, Lutgens E. Immune Modulation of Brown(ing) Adipose Tissue in Obesity. Endocr Rev 2017; 38:46-68. [PMID: 27849358 DOI: 10.1210/er.2016-1066] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 11/14/2016] [Indexed: 12/13/2022]
Abstract
Obesity is associated with a variety of medical conditions such as type 2 diabetes and cardiovascular diseases and is therefore responsible for high morbidity and mortality rates. Increasing energy expenditure by brown adipose tissue (BAT) is a current novel strategy to reduce the excessive energy stores in obesity. Brown adipocytes burn energy to generate heat and are mainly activated upon cold exposure. As prolonged cold exposure is not a realistic therapy, researchers worldwide are searching for novel ways to activate BAT and/or induce beiging of white adipose tissue. Recently, the contribution of immune cells in the regulation of brown adipocyte activity and beiging of white adipose tissue has gained increased attention, with a prominent role for eosinophils and alternatively activated macrophages. This review discusses the rediscovery of BAT, presents an overview of modes of activation and differentiation of beige and brown adipocytes, and describes the recently discovered immunological pathways that are key in mediating brown/beige adipocyte development and function. Interventions in immunological pathways harbor the potential to provide novel strategies to increase beige and brown adipose tissue activity as a therapeutic target for obesity.
Collapse
Affiliation(s)
- Susan M van den Berg
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Centre, University of Amsterdam, 1105AZ The Netherlands
| | - Andrea D van Dam
- Department of Medicine, Division of Endocrinology, and.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; and
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, and.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; and
| | - Menno P J de Winther
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Centre, University of Amsterdam, 1105AZ The Netherlands.,Institute for Cardiovascular Prevention, Ludwig Maximilians University of Munich, 80539 Munich, Germany
| | - Esther Lutgens
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Centre, University of Amsterdam, 1105AZ The Netherlands.,Institute for Cardiovascular Prevention, Ludwig Maximilians University of Munich, 80539 Munich, Germany
| |
Collapse
|
236
|
Sturla L, Mannino E, Scarfì S, Bruzzone S, Magnone M, Sociali G, Booz V, Guida L, Vigliarolo T, Fresia C, Emionite L, Buschiazzo A, Marini C, Sambuceti G, De Flora A, Zocchi E. Abscisic acid enhances glucose disposal and induces brown fat activity in adipocytes in vitro and in vivo. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:131-144. [PMID: 27871880 DOI: 10.1016/j.bbalip.2016.11.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 10/20/2016] [Accepted: 11/14/2016] [Indexed: 11/30/2022]
Abstract
Abscisic acid (ABA) is a plant hormone also present in animals, where it is involved in the regulation of innate immune cell function and of glucose disposal, through its receptor LANCL2. ABA stimulates glucose uptake by myocytes and pre-adipocytes in vitro and oral ABA improves glycemic control in rats and in healthy subjects. Here we investigated the role of the ABA/LANCL2 system in the regulation of glucose uptake and metabolism in adipocytes. Silencing of LANCL2 abrogated both the ABA- and insulin-induced increase of glucose transporter-4 expression and of glucose uptake in differentiated 3T3-L1 murine adipocytes; conversely, overexpression of LANCL2 enhanced basal, ABA- and insulin-stimulated glucose uptake. As compared with insulin, ABA treatment of adipocytes induced lower triglyceride accumulation, CO2 production and glucose-derived fatty acid synthesis. ABA per se did not induce pre-adipocyte differentiation in vitro, but stimulated adipocyte remodeling in terminally differentiated cells, with a reduction in cell size, increased mitochondrial content, enhanced O2 consumption, increased transcription of adiponectin and of brown adipose tissue (BAT) genes. A single dose of oral ABA (1μg/kg body weight) increased BAT glucose uptake 2-fold in treated rats compared with untreated controls. One-month-long ABA treatment at the same daily dose significantly upregulated expression of BAT markers in the WAT and in WAT-derived preadipocytes from treated mice compared with untreated controls. These results indicate a hitherto unknown role of LANCL2 in adipocyte sensitivity to insulin-stimulated glucose uptake and suggest a role for ABA in the induction and maintenance of BAT activity.
Collapse
Affiliation(s)
- Laura Sturla
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy.
| | - Elena Mannino
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Sonia Scarfì
- Department of Earth, Environment and Life Sciences, University of Genova, Via Pastore 3, 16132 Genova, Italy
| | - Santina Bruzzone
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Mirko Magnone
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Giovanna Sociali
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Valeria Booz
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Lucrezia Guida
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Tiziana Vigliarolo
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Chiara Fresia
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Laura Emionite
- Animal Facility, IRCCS AOU San Martino - IST, Genova, Italy
| | - Ambra Buschiazzo
- Nuclear Medicine, Dept of Health Sciences, University of Genova, Genova, Italy
| | - Cecilia Marini
- CNR Institute of Bioimages and Molecular Physiology, Section of Genova, Genova, Italy; IRCCS AOU San Martino - IST, Genova, Italy
| | - Gianmario Sambuceti
- Nuclear Medicine, Dept of Health Sciences, University of Genova, Genova, Italy; IRCCS AOU San Martino - IST, Genova, Italy
| | - Antonio De Flora
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Elena Zocchi
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| |
Collapse
|
237
|
Schatz V, Neubert P, Schröder A, Binger K, Gebhard M, Müller DN, Luft FC, Titze J, Jantsch J. Elementary immunology: Na + as a regulator of immunity. Pediatr Nephrol 2017; 32:201-210. [PMID: 26921211 PMCID: PMC5203836 DOI: 10.1007/s00467-016-3349-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 01/28/2016] [Accepted: 01/29/2016] [Indexed: 12/13/2022]
Abstract
The skin can serve as an interstitial Na+ reservoir. Local tissue Na+ accumulation increases with age, inflammation and infection. This increased local Na+ availability favors pro-inflammatory immune cell function and dampens their anti-inflammatory capacity. In this review, we summarize available data on how NaCl affects various immune cells. We particularly focus on how salt promotes pro-inflammatory macrophage and T cell function and simultaneously curtails their regulatory and anti-inflammatory potential. Overall, these findings demonstrate that local Na+ availability is a promising novel regulator of immunity. Hence, the modulation of tissue Na+ levels bears broad therapeutic potential: increasing local Na+ availability may help in treating infections, while lowering tissue Na+ levels may be used to treat, for example, autoimmune and cardiovascular diseases.
Collapse
Affiliation(s)
- Valentin Schatz
- Institute of Clinical Microbiology and Hygiene, Universitätsklinikum Regensburg-Universität Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Patrick Neubert
- Institute of Clinical Microbiology and Hygiene, Universitätsklinikum Regensburg-Universität Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Agnes Schröder
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen-Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Katrina Binger
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Matthias Gebhard
- Experimental and Clinical Research Center (ECRC), Research Building, Charité Lindenberger Weg 80, Berlin, Germany
- Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Dominik N Müller
- Experimental and Clinical Research Center (ECRC), Research Building, Charité Lindenberger Weg 80, Berlin, Germany
- Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Friedrich C Luft
- Experimental and Clinical Research Center (ECRC), Research Building, Charité Lindenberger Weg 80, Berlin, Germany
- Max-Delbrück Center for Molecular Medicine, Berlin, Germany
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jens Titze
- Department of Nephrology and Hypertension, Universitätsklinikum Erlangen-Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jonathan Jantsch
- Institute of Clinical Microbiology and Hygiene, Universitätsklinikum Regensburg-Universität Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany.
| |
Collapse
|
238
|
Shu L, Hoo RLC, Wu X, Pan Y, Lee IPC, Cheong LY, Bornstein SR, Rong X, Guo J, Xu A. A-FABP mediates adaptive thermogenesis by promoting intracellular activation of thyroid hormones in brown adipocytes. Nat Commun 2017; 8:14147. [PMID: 28128199 PMCID: PMC5290165 DOI: 10.1038/ncomms14147] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 12/02/2016] [Indexed: 01/01/2023] Open
Abstract
The adipokine adipocyte fatty acid-binding protein (A-FABP) has been implicated in obesity-related cardio-metabolic complications. Here we show that A-FABP increases thermogenesis by promoting the conversion of T4 to T3 in brown adipocytes. We find that A-FABP levels are increased in both white (WAT) and brown (BAT) adipose tissues and the bloodstream in response to thermogenic stimuli. A-FABP knockout mice have reduced thermogenesis and whole-body energy expenditure after cold stress or after feeding a high-fat diet, which can be reversed by infusion of recombinant A-FABP. Mechanistically, A-FABP induces the expression of type-II iodothyronine deiodinase in BAT via inhibition of the nuclear receptor liver X receptor α, thereby leading to the conversion of thyroid hormone from its inactive form T4 to active T3. The thermogenic responses to T4 are abrogated in A-FABP KO mice, but enhanced by A-FABP. Thus, A-FABP acts as a physiological stimulator of BAT-mediated adaptive thermogenesis.
Collapse
Affiliation(s)
- Lingling Shu
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ruby L. C. Hoo
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xiaoping Wu
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yong Pan
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ida P. C. Lee
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Lai Yee Cheong
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | | | - Xianglu Rong
- Joint Laboratory of Guangdong and Hong Kong on Metabolic Diseases, Guangdong Pharmaceutical University, 510000 Guangzhou, China
| | - Jiao Guo
- Joint Laboratory of Guangdong and Hong Kong on Metabolic Diseases, Guangdong Pharmaceutical University, 510000 Guangzhou, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
239
|
Saito N, Kimura S, Miyamoto T, Fukushima S, Amagasa M, Shimamoto Y, Nishioka C, Okamoto S, Toda C, Washio K, Asano A, Miyoshi I, Takahashi E, Kitamura H. Macrophage ubiquitin-specific protease 2 modifies insulin sensitivity in obese mice. Biochem Biophys Rep 2017; 9:322-329. [PMID: 28956020 PMCID: PMC5614627 DOI: 10.1016/j.bbrep.2017.01.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 12/24/2016] [Accepted: 01/23/2017] [Indexed: 12/17/2022] Open
Abstract
We previously reported that ubiquitin-specific protease (USP) 2 in macrophages down-regulates genes associated with metabolic diseases, suggesting a putative anti-diabetic role for USP2 in macrophages. In this study, we evaluate this role at both cellular and individual levels. Isolated macrophages forcibly expressing Usp2a, a longer splicing variant of USP2, failed to modulate the insulin sensitivity of 3T3-L1 adipocytes. Similarly, macrophage-selective overexpression of Usp2a in mice (Usp2a transgenic mice) had a negligible effect on insulin sensitivity relative to wild type littermates following a three-month high-fat diet. However, Usp2a transgenic mice exhibited fewer M1 macrophages in their mesenteric adipose tissue. Following a six-month high-fat diet, Usp2a transgenic mice exhibited a retarded progression of insulin resistance in their skeletal muscle and liver, and an improvement in insulin sensitivity at an individual level. Although conditioned media from Usp2a-overexpressing macrophages did not directly affect the insulin sensitivity of C2C12 myotubes compared to media from control macrophages, they did increase the insulin sensitivity of C2C12 cells after subsequent conditioning with 3T3-L1 cells. These results indicate that macrophage USP2A hampers obesity-elicited insulin resistance via an adipocyte-dependent mechanism. USP2A controls macrophage population in mesenteric adipose tissue during obesity. Overexpression of USP2A in macrophages retards progression of insulin resistance. Overexpression of USP2A in macrophages represses high-fat diet-induced obesity. Macrophage USP2A controls insulin sensitivity of muscle dependent on adipocytes.
Collapse
Key Words
- DMEM, Dulbecco's modified Eagle medium
- Diabetes
- ELISA, enzyme-linked immunosorbent assay
- GAPDH, glyceraldehyde 3-phosphate dehydrogenase
- HFD, high-fat diet
- HOMA-IR, homeostatic model assessment as an index of insulin resistance
- IL, interleukin
- IR, insulin receptor
- IRS, insulin receptor substrate
- Insulin
- KD, knock down
- KO, knockout
- Macrophage
- NCD, normal chow diet
- NEFA, nonesterified fatty acid
- Obesity
- PDK, phosphoinositide-dependent kinase
- PI3K, phosphatidylinositol 3-phosphate kinase
- SOCS, suppressor of cytokine signaling
- T2DM, type 2 diabetes mellitus
- Tg, transgenic
- USP
- USP, ubiquitin-specific protease
- pAkt, phosphorylated Akt
- pIRβ, phosphorylated insulin receptor β chain
Collapse
Affiliation(s)
- Natsuko Saito
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, and Laboratory of Animal Therapeutics, Japan
| | - Shunsuke Kimura
- Laboratory of Histology and Cytology, Department of Functional Morphology, Graduate School of Medical Sciences, Hokkaido University, Kita15, Nishi7, Kita-ku, Sapporo, Hokkaido 060-8638, Japan
| | - Tomomi Miyamoto
- Department of Comparative and Experimental Medicine, Graduate School of Medicine, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Sanae Fukushima
- Research Resources Center, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Misato Amagasa
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, and Laboratory of Animal Therapeutics, Japan
| | - Yoshinori Shimamoto
- Department of Veterinary Science, Rakuno Gakuen University, 582 Midorimachi, Bunkyodai, Ebetsu, Hokkaido 069-8501, Japan
| | - Chieko Nishioka
- Research Resources Center, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Shiki Okamoto
- Division of Endocrinology and Metabolism, National Institute for Physiological Sciences, 38 Nishigonaka Myodaiji, Okazaki, Aichi 444-8585, Japan
| | - Chitoku Toda
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8063, USA
| | - Kohei Washio
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, and Laboratory of Animal Therapeutics, Japan
| | - Atsushi Asano
- Laboratory of Laboratory Animal, Department of Basic Veterinary Science, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Ichiro Miyoshi
- Department of Comparative and Experimental Medicine, Graduate School of Medicine, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| | - Eiki Takahashi
- Research Resources Center, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Hiroshi Kitamura
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, and Laboratory of Animal Therapeutics, Japan.,Laboratory of Histology and Cytology, Department of Functional Morphology, Graduate School of Medical Sciences, Hokkaido University, Kita15, Nishi7, Kita-ku, Sapporo, Hokkaido 060-8638, Japan.,Department of Comparative and Experimental Medicine, Graduate School of Medicine, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi 467-8601, Japan
| |
Collapse
|
240
|
Man K, Kutyavin VI, Chawla A. Tissue Immunometabolism: Development, Physiology, and Pathobiology. Cell Metab 2017; 25:11-26. [PMID: 27693378 PMCID: PMC5226870 DOI: 10.1016/j.cmet.2016.08.016] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/15/2016] [Accepted: 08/24/2016] [Indexed: 02/07/2023]
Abstract
Evolution of metazoans resulted in the specialization of cellular and tissue function. This was accomplished by division of labor, which allowed tissue parenchymal cells to prioritize their core functions while ancillary functions were delegated to tissue accessory cells, such as immune, stromal, and endothelial cells. In metabolic organs, the accessory cells communicate with their clients, the tissue parenchymal cells, to optimize cellular processes, allowing organisms to adapt to changes in their environment. Here, we discuss tissue immunometabolism from this vantage point and use examples from adipose tissues (white, beige, and brown) and liver to outline the general principles by which accessory cells support metabolic homeostasis in parenchymal cells. A corollary of this model is that disruption of communication between client and accessory cells might predispose metabolic organs to the development of disease.
Collapse
Affiliation(s)
- Kevin Man
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-0795, USA
| | - Vassily I Kutyavin
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-0795, USA
| | - Ajay Chawla
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-0795, USA; Departments of Physiology and Medicine, University of California, San Francisco, CA 94143-0795, USA.
| |
Collapse
|
241
|
Abstract
Brown adipose tissue (BAT) is the main site of adaptive thermogenesis and experimental studies have associated BAT activity with protection against obesity and metabolic diseases, such as type 2 diabetes mellitus and dyslipidaemia. Active BAT is present in adult humans and its activity is impaired in patients with obesity. The ability of BAT to protect against chronic metabolic disease has traditionally been attributed to its capacity to utilize glucose and lipids for thermogenesis. However, BAT might also have a secretory role, which could contribute to the systemic consequences of BAT activity. Several BAT-derived molecules that act in a paracrine or autocrine manner have been identified. Most of these factors promote hypertrophy and hyperplasia of BAT, vascularization, innervation and blood flow, processes that are all associated with BAT recruitment when thermogenic activity is enhanced. Additionally, BAT can release regulatory molecules that act on other tissues and organs. This secretory capacity of BAT is thought to be involved in the beneficial effects of BAT transplantation in rodents. Fibroblast growth factor 21, IL-6 and neuregulin 4 are among the first BAT-derived endocrine factors to be identified. In this Review, we discuss the current understanding of the regulatory molecules (the so-called brown adipokines or batokines) that are released by BAT that influence systemic metabolism and convey the beneficial metabolic effects of BAT activation. The identification of such adipokines might also direct drug discovery approaches for managing obesity and its associated chronic metabolic diseases.
Collapse
Affiliation(s)
- Francesc Villarroya
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina, Universitat de Barcelona, Avda Diagonal 643, 08028-Barcelona, Catalonia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Facultat de Biologia, Universitat de Barcelona, Avda Diagonal 643, 08028-Barcelona, Catalonia, Spain
| | - Rubén Cereijo
- CIBER Fisiopatología de la Obesidad y Nutrición, Facultat de Biologia, Universitat de Barcelona, Avda Diagonal 643, 08028-Barcelona, Catalonia, Spain
| | - Joan Villarroya
- CIBER Fisiopatología de la Obesidad y Nutrición, Facultat de Biologia, Universitat de Barcelona, Avda Diagonal 643, 08028-Barcelona, Catalonia, Spain
| | - Marta Giralt
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina, Universitat de Barcelona, Avda Diagonal 643, 08028-Barcelona, Catalonia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Facultat de Biologia, Universitat de Barcelona, Avda Diagonal 643, 08028-Barcelona, Catalonia, Spain
| |
Collapse
|
242
|
Oh KJ, Lee DS, Kim WK, Han BS, Lee SC, Bae KH. Metabolic Adaptation in Obesity and Type II Diabetes: Myokines, Adipokines and Hepatokines. Int J Mol Sci 2016; 18:ijms18010008. [PMID: 28025491 PMCID: PMC5297643 DOI: 10.3390/ijms18010008] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/24/2016] [Accepted: 12/12/2016] [Indexed: 12/21/2022] Open
Abstract
Obesity and type II diabetes are characterized by insulin resistance in peripheral tissues. A high caloric intake combined with a sedentary lifestyle is the leading cause of these conditions. Whole-body insulin resistance and its improvement are the result of the combined actions of each insulin-sensitive organ. Among the fundamental molecular mechanisms by which each organ is able to communicate and engage in cross-talk are cytokines or peptides which stem from secretory organs. Recently, it was reported that several cytokines or peptides are secreted from muscle (myokines), adipose tissue (adipokines) and liver (hepatokines) in response to certain nutrition and/or physical activity conditions. Cytokines exert autocrine, paracrine or endocrine effects for the maintenance of energy homeostasis. The present review is focused on the relationship and cross-talk amongst muscle, adipose tissue and the liver as secretory organs in metabolic diseases.
Collapse
Affiliation(s)
- Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea.
| | - Da Som Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
| | - Won Kon Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea.
| | - Baek Soo Han
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea.
| | - Sang Chul Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea.
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea.
| |
Collapse
|
243
|
Abstract
Adipose tissue plays a central role in regulating whole-body energy and glucose homeostasis through its subtle functions at both organ and systemic levels. On one hand, adipose tissue stores energy in the form of lipid and controls the lipid mobilization and distribution in the body. On the other hand, adipose tissue acts as an endocrine organ and produces numerous bioactive factors such as adipokines that communicate with other organs and modulate a range of metabolic pathways. Moreover, brown and beige adipose tissue burn lipid by dissipating energy in the form of heat to maintain euthermia, and have been considered as a new way to counteract obesity. Therefore, adipose tissue dysfunction plays a prominent role in the development of obesity and its related disorders such as insulin resistance, cardiovascular disease, diabetes, depression and cancer. In this review, we will summarize the recent findings of adipose tissue in the control of metabolism, focusing on its endocrine and thermogenic function.
Collapse
Affiliation(s)
- Liping Luo
- Department of Metabolism and EndocrinologyMetabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Meilian Liu
- Department of Metabolism and EndocrinologyMetabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Department of Biochemistry and Molecular BiologyUniversity of New Mexico Health Sciences Center,
Albuquerque, New Mexico, USA
| |
Collapse
|
244
|
Adipose tissue macrophage in immune regulation of metabolism. SCIENCE CHINA-LIFE SCIENCES 2016; 59:1232-1240. [DOI: 10.1007/s11427-016-0155-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 09/13/2016] [Indexed: 12/14/2022]
|
245
|
Gender-Specific Mechanisms Underlying the Amelioration of High-Fat Diet-Induced Glucose Intolerance in B-Cell-Activating Factor Deficient Mice. PLoS One 2016; 11:e0166225. [PMID: 27814392 PMCID: PMC5096712 DOI: 10.1371/journal.pone.0166225] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 10/12/2016] [Indexed: 01/23/2023] Open
Abstract
It has recently been found that B cell activating factor (BAFF) plays an important role in the regulation of energy homeostasis. We also have previously reported that BAFF deficiency reverses high-fat (HF) diet-induced glucose intolerance by potentiating adipose tissue function. In the present study, we found that BAFF deficient (BAFF-/-) mice exhibit gender-specific differences in protection against diet-induced glucose intolerance, and aimed to characterize the gender-dependent molecular alterations in energy metabolism. Under HF feeding conditions, serum BAFF level of female wild-type (WT) mice was considerably higher than that of male mice. Despite increased body weight gain, both male and female BAFF-/- mice showed significantly improved glucose tolerance compared to their WT counterparts. Expressions of genes involved in glucose transport, thermogenesis and lipid oxidation were up-regulated in brown adipose tissues of both male and female BAFF-/- mice. Interestingly, the expression of thermogenic genes in subcutaneous adipose tissue was significantly enhanced in female BAFF-/- compared to WT mice, but the difference was not observed between male BAFF-/- and WT mice. The enhanced thermogenic program was confirmed by higher protein levels of UCP1 and irisin in female BAFF-/- than in WT mice. Additionally, adiponectin production in white adipose tissues and AMPK phosphorylation in subcutaneous adipose tissue were also significantly elevated in female BAFF-/- compared to WT mice, but not in male BAFF-/- mice. Our findings define a comprehensive scenario for the enhancing effect of BAFF depletion on glucose tolerance wherein the underlying mechanism is, at least in part, gender-specific, and suggest that gender difference should be considered as an important factor in the use of BAFF blockade as a therapeutic approach for the prevention and treatment of type 2 diabetes.
Collapse
|
246
|
Abstract
Adipose tissue not only functions as the major energy-storing tissue, but also functions as an endocrine organ that regulates systemic metabolism by releasing various hormones called adipokines. Macrophages play a critical role in maintaining adipocyte health in a lean state and in remodeling during the progression of obesity. Large numbers of classically activated (M1) macrophages accumulate in adipose tissue as adipocytes become larger because of excessive energy conditions, and they adversely affect insulin resistance by triggering local and systemic inflammation. In contrast, alternatively activated (M2) macrophages seem to maintain the health of adipose tissues in a lean state. In addition, they play a role in adapting to excess energy states, because M2 macrophage dysfunction caused by genetic disruption of the M2 gene results in metabolic disorders under high-fat-fed conditions that are probably attributable to their anti-inflammatory functions. Nonetheless, how M2 macrophages contribute to maintaining the health of adipose tissue and therefore to insulin sensitivity is largely unknown. In this article, we review the literature on the role of M1 and M2 macrophages in metabolism, with a special focus on the role of M2 macrophages in adipose tissue. Likewise, we raise topics of M2 macrophages in non-adipose tissues to expand our understanding of macrophage heterogeneity.
Collapse
|
247
|
Metabolic control of immune tolerance in health and autoimmunity. Semin Immunol 2016; 28:491-504. [PMID: 27720234 DOI: 10.1016/j.smim.2016.09.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/23/2016] [Accepted: 09/30/2016] [Indexed: 12/15/2022]
Abstract
The filed that links immunity and metabolism is rapidly expanding. The adipose tissue, by secreting a series of immune regulators called adipokines, represents the common mediator linking metabolic processes and immune system functions. The dysregulation of adipokine secretion, occurring in obese individuals or in conditions of malnutrition or dietary restriction, affects the activity of immune cells resulting in inflammatory autoimmune responses or increased susceptibility to infectious diseases. Alterations of cell metabolism that characterize several autoimmune diseases strongly support the idea that the immune tolerance is also regulated by metabolic pathways. The comprehension of the molecular mechanisms underlying these alterations may lead to the development of novel therapeutic strategies to control immune cell differentiation and function in conditions of autoimmunity.
Collapse
|
248
|
Zhu Q, Ghoshal S, Rodrigues A, Gao S, Asterian A, Kamenecka TM, Barrow JC, Chakraborty A. Adipocyte-specific deletion of Ip6k1 reduces diet-induced obesity by enhancing AMPK-mediated thermogenesis. J Clin Invest 2016; 126:4273-4288. [PMID: 27701146 DOI: 10.1172/jci85510] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 08/29/2016] [Indexed: 12/15/2022] Open
Abstract
Enhancing energy expenditure (EE) is an attractive strategy to combat obesity and diabetes. Global deletion of Ip6k1 protects mice from diet-induced obesity (DIO) and insulin resistance, but the tissue-specific mechanism by which IP6K1 regulates body weight is unknown. Here, we have demonstrated that IP6K1 regulates fat accumulation by modulating AMPK-mediated adipocyte energy metabolism. Cold exposure led to downregulation of Ip6k1 in murine inguinal and retroperitoneal white adipose tissue (IWAT and RWAT) depots. Adipocyte-specific deletion of Ip6k1 (AdKO) enhanced thermogenic EE, which protected mice from high-fat diet-induced weight gain at ambient temperature (23°C), but not at thermoneutral temperature (30°C). AdKO-induced increases in thermogenesis also protected mice from cold-induced decreases in body temperature. UCP1, PGC1α, and other markers of browning and thermogenesis were elevated in IWAT and RWAT of AdKO mice. Cold-induced activation of sympathetic signaling was unaltered, whereas AMPK was enhanced, in AdKO IWAT. Moreover, beige adipocytes from AdKO IWAT displayed enhanced browning, which was diminished by AMPK depletion. Furthermore, we determined that IP6 and IP6K1 differentially regulate upstream kinase-mediated AMPK stimulatory phosphorylation in vitro. Finally, treating mildly obese mice with the IP6K inhibitor TNP enhanced thermogenesis and inhibited progression of DIO. Thus, IP6K1 regulates energy metabolism via a mechanism that could potentially be targeted in obesity.
Collapse
|
249
|
The modern interleukin-1 superfamily: Divergent roles in obesity. Semin Immunol 2016; 28:441-449. [DOI: 10.1016/j.smim.2016.10.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/03/2016] [Accepted: 10/03/2016] [Indexed: 11/20/2022]
|
250
|
Lv Y, Zhang SY, Liang X, Zhang H, Xu Z, Liu B, Xu MJ, Jiang C, Shang J, Wang X. Adrenomedullin 2 Enhances Beiging in White Adipose Tissue Directly in an Adipocyte-autonomous Manner and Indirectly through Activation of M2 Macrophages. J Biol Chem 2016; 291:23390-23402. [PMID: 27621315 DOI: 10.1074/jbc.m116.735563] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Indexed: 12/15/2022] Open
Abstract
Adrenomedullin 2 (ADM2) is an endogenous bioactive peptide belonging to the calcitonin gene-related peptide family. Our previous studies showed that overexpression of ADM2 in mice reduced obesity and insulin resistance by increasing thermogenesis in brown adipose tissue. However, the effects of ADM2 in another type of thermogenic adipocyte, beige adipocytes, remain to be understood. The plasma ADM2 levels were inversely correlated with obesity in humans, and adipo-ADM2-transgenic (tg) mice displayed resistance to high-fat diet-induced obesity with increased energy expenditure. Beiging of subcutaneous white adipose tissues (WAT) was more noticeably induced in high-fat diet-fed transgenic mice with adipocyte-ADM2 overexpression (adipo-ADM2-tg mice) than in WT animals. ADM2 treatment in primary rat subcutaneous adipocytes induced beiging with up-regulation of UCP1 and beiging-related marker genes and increased mitochondrial uncoupling respiration, which was mainly mediated by activation of the calcitonin receptor-like receptor (CRLR)·receptor activity-modifying protein 1 (RAMP1) complex and PKA and p38 MAPK signaling pathways. Importantly, this adipocyte-autonomous beiging effect by ADM2 was translatable to human primary adipocytes. In addition, M2 macrophage activation also contributed to the beiging effects of ADM2 through catecholamine secretion. Therefore, our study reveals that ADM2 enhances subcutaneous WAT beiging via a direct effect by activating the CRLR·RAMP1-cAMP/PKA and p38 MAPK pathways in white adipocytes and via an indirect effect by stimulating alternative M2 polarization in macrophages. Through both mechanisms, beiging of WAT by ADM2 results in increased energy expenditure and reduced obesity, suggesting ADM2 as a novel anti-obesity target.
Collapse
Affiliation(s)
- Ying Lv
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Song-Yang Zhang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Xianyi Liang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Heng Zhang
- the Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Zhi Xu
- the Department of General Surgery, Peking University Third Hospital, Beijing 100191, China, and
| | - Bo Liu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Ming-Jiang Xu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Changtao Jiang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China,
| | - Jin Shang
- the Department of Cardiometabolic Disease, Merck Research Laboratories, Merck & Co, Inc., Kenilworth, New Jersey 07033
| | - Xian Wang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| |
Collapse
|