401
|
Cheung WW, Cherqui S, Ding W, Esparza M, Zhou P, Shao J, Lieber RL, Mak RH. Muscle wasting and adipose tissue browning in infantile nephropathic cystinosis. J Cachexia Sarcopenia Muscle 2016; 7:152-64. [PMID: 27493869 PMCID: PMC4864942 DOI: 10.1002/jcsm.12056] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 04/21/2015] [Accepted: 06/03/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Muscle wasting is a common complication in patients with infantile nephropathic cystinosis, but its mechanism and association with energy metabolism is not known. We define the metabolic phenotype in Ctns(-/-) mice, an established murine model of infantile nephropathic cystinosis, with focus on muscle wasting and energy homeostasis. METHODS Male Ctns(-/-) mice and wild-type (WT) controls were studied at 1, 4, 9, and 12 months of age. As Ctns(-/-) mice started to develop chronic kidney disease (CKD) at 9 months of age, 9- and 12-month-old Ctns(-/-) mice were also compared with age-matched WT mice with CKD. Serum and urine chemistry and energy homeostasis parameters were measured. Skeletal muscle histomorphometry and in vivo muscle function were measured. We studied expression of genes involved in muscle mass regulation, thermogenesis, energy metabolism, adipogenesis, and adipose tissue browning in Ctns(-/-) mice. RESULTS Ctns(-/-) mice showed loss of weight and lean mass and increased energy expenditure. Ctns(-/-) mice exhibited abnormal energy homeostasis before the onset of their CKD. Food intake in Ctns(-/-) mice was comparable with age-matched WT controls. However, significantly lower total body mass starting at 1 month of age and increased energy expenditure at 4 months of age preceded the onset of CKD at 9 months of age in Ctns(-/-) mice. Muscle accept content in 1- and 4-month-old Ctns(-/-) mice was significantly lower than that in age-matched WT controls. At 12 months of age, muscle fibre area and in vivo muscle strength was reduced in Ctns(-/-) mice than that in WT or CKD controls. Muscle wasting in Ctns(-/-) mice was associated with inhibition of myogenesis, activation of muscle proteolysis pathways, and overexpression of pro-inflammatory cytokines. Increased energy expenditure was associated with elevation of thermogenesis in skeletal muscle and adipose tissues. The development of beige adipocytes in Ctns(-/-) mice is a novel finding. Expression of beige adipose cell surface markers (CD137, Tmem26, and Tbx1) and uncoupling protein-1, which is a brown adipose tissue marker, was observed in inguinal white adipose tissue of Ctns(-/-) mice. Expression of key molecules implicated in the pathogenesis of adipose tissue browning (Cox2, cytochrome c oxidase subunit II; PGF2α, prostaglandin F2α; IL-1α, interleukin 1α; IL-6, interleukin 6; TNF-α, tumor necrosis factor α) was significantly increased in inguinal white adipose tissue of Ctns(-/-) mice than that in WT controls. CONCLUSION This study describes a mouse model of nephropathic cystinosis presenting with profound muscle wasting. The mechanism for hypermetabolism in Ctns(-/-) mice may involve up-regulation of thermogenesis pathways in skeletal muscle and adipose tissues. This study demonstrates, for the first time, the development of beige adipocytes in Ctns(-/-) mice. Understanding the underlying mechanisms of adipose tissue browning in cystinosis may lead to novel therapy.
Collapse
Affiliation(s)
- Wai W Cheung
- Department of Pediatrics University of California San Diego CA USA
| | | | - Wei Ding
- Department of Pediatrics University of California San Diego CA USA; Division of Nephrology, The 5th People's Hospital of Shanghai Fudan University Shanghai China
| | - Mary Esparza
- Department of Orthopedic Surgery University of California San Diego CA USA
| | - Ping Zhou
- Department of Pediatrics University of California San Diego CA USA; Department of Pediatrics The 2nd Hospital of Harbin Medical University Harbin China
| | - Jianhua Shao
- Department of Pediatrics University of California San Diego CA USA
| | - Richard L Lieber
- Department of Orthopedic Surgery University of California San Diego CA USA; Rehabilitation Institute of Chicago Chicago
| | - Robert H Mak
- Department of Pediatrics University of California San Diego CA USA
| |
Collapse
|
402
|
Exosomal microRNA miR-92a concentration in serum reflects human brown fat activity. Nat Commun 2016; 7:11420. [PMID: 27117818 PMCID: PMC4853423 DOI: 10.1038/ncomms11420] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 03/24/2016] [Indexed: 12/29/2022] Open
Abstract
Brown adipose tissue (BAT) dissipates energy and its activity correlates with leanness in human adults. 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography coupled with computer tomography (PET/CT) is still the standard for measuring BAT activity, but exposes subjects to ionizing radiation. To study BAT function in large human cohorts, novel diagnostic tools are needed. Here we show that brown adipocytes release exosomes and that BAT activation increases exosome release. Profiling miRNAs in exosomes released from brown adipocytes, and in exosomes isolated from mouse serum, we show that levels of miRNAs change after BAT activation in vitro and in vivo. One of these exosomal miRNAs, miR-92a, is also present in human serum exosomes. Importantly, serum concentrations of exosomal miR-92a inversely correlate with human BAT activity measured by 18F-FDG PET/CT in two unique and independent cohorts comprising 41 healthy individuals. Thus, exosomal miR-92a represents a potential serum biomarker for BAT activity in mice and humans. Exosomes are RNA-containing lipid vesicles with roles in inter-tissue crosstalk. Here the authors show that exosome release from brown adipocytes is increased upon thermogenic activation, both in vitro and in vivo, and demonstrate that serum levels of exosomal miR-92 reflect brown fat activity in humans.
Collapse
|
403
|
Lo KA, Ng PY, Kabiri Z, Virshup D, Sun L. Wnt inhibition enhances browning of mouse primary white adipocytes. Adipocyte 2016; 5:224-31. [PMID: 27386162 PMCID: PMC4916886 DOI: 10.1080/21623945.2016.1148834] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 01/18/2016] [Accepted: 01/20/2016] [Indexed: 01/01/2023] Open
Abstract
The global epidemic in obesity and metabolic syndrome requires novel approaches to tackle. White adipose tissue, traditionally seen as a passive energy-storage organ, can be induced to take on certain characteristics of brown fat in a process called browning. The “browned” white adipose tissue, or beige fat, is a potential anti-obesity target. Various signaling pathways can enhance browning. Wnt is a key regulator of adipocyte biology, but its role in browning has not been explored. In this study, we found that in primary mouse adipocytes derived from the inguinal depot, Wnt inhibition by both chemical and genetic methods significantly enhanced browning. The effect of Wnt inhibition on browning most likely targets the beige precursor cells in selected adipose depots.
Collapse
Affiliation(s)
- Kinyui Alice Lo
- Programme in Cardiovascular & Metabolic Disorders, Duke-NUS, Singapore
- Institute of Molecular and Cell Biology, Proteos, Singapore
| | - Pei Yi Ng
- Institute of Molecular and Cell Biology, Proteos, Singapore
| | - Zahra Kabiri
- Programme in Cancer and Stem Cell Biology, Duke-NUS, Singapore
| | - David Virshup
- Programme in Cancer and Stem Cell Biology, Duke-NUS, Singapore
| | - Lei Sun
- Programme in Cardiovascular & Metabolic Disorders, Duke-NUS, Singapore
- Institute of Molecular and Cell Biology, Proteos, Singapore
| |
Collapse
|
404
|
Longchamp A, Tao M, Bartelt A, Ding K, Lynch L, Hine C, Corpataux JM, Kristal BS, Mitchell JR, Ozaki CK. Surgical injury induces local and distant adipose tissue browning. Adipocyte 2016; 5:163-74. [PMID: 27386152 DOI: 10.1080/21623945.2015.1111971] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 10/12/2015] [Accepted: 10/16/2015] [Indexed: 12/20/2022] Open
Abstract
The adipose organ, which comprises brown, white and beige adipocytes, possesses remarkable plasticity in response to feeding and cold exposure. The development of beige adipocytes in white adipose tissue (WAT), a process called browning, represents a promising route to treat metabolic disorders. While surgical procedures constantly traumatize adipose tissue, its impact on adipocyte phenotype remains to be established. Herein, we studied the effect of trauma on adipocyte phenotype one day after sham, incision control, or surgical injury to the left inguinal adipose compartment. Caloric restriction was used to control for surgery-associated body temperature changes and weight loss. We characterized the trauma-induced cellular and molecular changes in subcutaneous, visceral, interscapular, and perivascular adipose tissue using histology, immunohistochemistry, gene expression, and flow cytometry analysis. After one day, surgical trauma stimulated adipose tissue browning at the site of injury and, importantly, in the contralateral inguinal depot. Browning was not present after incision only, and was largely independent of surgery-associated body temperature and weight loss. Adipose trauma rapidly recruited monocytes to the injured site and promoted alternatively activated macrophages. Conversely, PDGF receptor-positive beige progenitors were reduced. In this study, we identify adipose trauma as an unexpected driver of selected local and remote adipose tissue browning, holding important implications for the biologic response to surgical injury.
Collapse
Affiliation(s)
- Alban Longchamp
- Department of Surgery and the Heart and Vascular Center, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| | - Ming Tao
- Department of Surgery and the Heart and Vascular Center, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| | - Alexander Bartelt
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, MA, USA
| | - Kui Ding
- Department of Surgery and the Heart and Vascular Center, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| | - Lydia Lynch
- Department of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| | - Christopher Hine
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, MA, USA
| | - Jean-Marc Corpataux
- Department of Thoracic and Vascular Surgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Bruce S. Kristal
- Department of Neurosurgery, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| | - James R. Mitchell
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, MA, USA
| | - C. Keith Ozaki
- Department of Surgery and the Heart and Vascular Center, Brigham and Women's Hospital/Harvard Medical School, Boston, MA, USA
| |
Collapse
|
405
|
Bai Y, Shang Q, Zhao H, Pan Z, Guo C, Zhang L, Wang Q. Pdcd4 restrains the self-renewal and white-to-beige transdifferentiation of adipose-derived stem cells. Cell Death Dis 2016; 7:e2169. [PMID: 27031966 PMCID: PMC4823969 DOI: 10.1038/cddis.2016.75] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 02/23/2016] [Accepted: 03/03/2016] [Indexed: 12/31/2022]
Abstract
The stemness maintenance of adipose-derived stem cells (ADSCs) is important for adipose homeostasis and energy balance. Programmed cell death 4 (Pdcd4) has been demonstrated to be involved in the development of obesity, but its possible roles in ADSC function and adipogenic capacity remain unclear. In this study, we demonstrate that Pdcd4 is a key controller that limits the self-renewal and white-to-beige transdifferentiation of ADSCs. Pdcd4 deficiency in mice caused stemness enhancement of ADSCs as evidenced by increased expression of CD105, CD90, Nanog and Oct4 on ADSCs, together with enhanced in situ proliferation in adipose tissues. Pdcd4 deficiency promoted proliferation, colony formation of ADSCs and drove more ADSCs entering the S phase accompanied by AKT activation and cyclinD1 upregulation. Blockade of AKT signaling in Pdcd4-deficient ADSCs led to a marked decline in cyclinD1, S-phase entry and cell proliferation, revealing AKT as a target for repressing ADSC self-renewal by Pdcd4. Intriguingly, depletion of Pdcd4 promoted the transdifferentiation of ADSCs into beige adipocytes. A reduction in lipid contents and expression levels of white adipocyte markers including C/EBPα, PPAR-γ, adiponectin and αP2 was detected in Pdcd4-deficient ADSCs during white adipogenic differentiation, substituted by typical beige adipocyte characteristics including small, multilocular lipid droplets and UCP1 expression. More lactate produced by Pdcd4-deficient ADSCs might be an important contributor to the expression of UCP1 and white-to-beige transdifferentiation. In addition, an elevation of UCP1 expression was confirmed in white adipose tissues from Pdcd4-deficient mice upon high-fat diet, which displayed increased energy expenditure and resistance to obesity as compared with wild-type obese mice. These findings provide evidences that Pdcd4 produces unfavorable influences on ADSC stemness, which contribute to adipose dysfunction, obesity and metabolic syndromes, thereby proposing Pdcd4 as a potential intervening target for regulating ADSC function.
Collapse
Affiliation(s)
- Y Bai
- Department of Immunology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Q Shang
- Department of Immunology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - H Zhao
- Department of Immunology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Z Pan
- Department of Immunology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - C Guo
- Department of Immunology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - L Zhang
- Department of Immunology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Q Wang
- Department of Immunology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| |
Collapse
|
406
|
Suchacki KJ, Cawthorn WP, Rosen CJ. Bone marrow adipose tissue: formation, function and regulation. Curr Opin Pharmacol 2016; 28:50-6. [PMID: 27022859 DOI: 10.1016/j.coph.2016.03.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 03/11/2016] [Accepted: 03/11/2016] [Indexed: 11/27/2022]
Abstract
The human body requires an uninterrupted supply of energy to maintain metabolic homeostasis and energy balance. To sustain energy balance, excess consumed calories are stored as glycogen, triglycerides and protein, allowing the body to continue to function in states of starvation and increased energy expenditure. Adipose tissue provides the largest natural store of excess calories as triglycerides and plays an important role as an endocrine organ in energy homeostasis and beyond. This short review is intended to detail the current knowledge of the formation and role of bone marrow adipose tissue (MAT), a largely ignored adipose depot, focussing on the role of MAT as an endocrine organ and highlighting the pharmacological agents that regulate MAT.
Collapse
Affiliation(s)
- Karla J Suchacki
- The Queen's Medical Research Institute, University of Edinburgh, UK.
| | | | | |
Collapse
|
407
|
Kalinovich AV, Shabalina IG. Novel Mitochondrial Cationic Uncoupler C4R1 Is an Effective Treatment for Combating Obesity in Mice. BIOCHEMISTRY (MOSCOW) 2016; 80:620-8. [PMID: 26071782 DOI: 10.1134/s0006297915050156] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Obesity is associated with premature mortality, impaired quality of life, and large healthcare costs. However, treatment options remain quite limited. Here we studied potential anti-obesity effects of a novel cationic mitochondrial uncoupler, C4R1 (derivative of rhodamine 19) in C57Bl/6 mice. Obesity was induced by long-term (eight weeks) high fat diet feeding at thermoneutrality. The treated group of mice received consecutively two doses of C4R1 in drinking water (30 and 12-14 µmol/kg daily) during 30 days. Effects of C4R1 were dose-dependent. After six days of C4R1 treatment at dose 30 µmol/kg daily, food intake was reduced by 68%, body weight by 19%, and fat mass by 21%. Body weight decrease was explained partly by reduced food intake and partly by increased metabolism, likely resulting from uncoupling. Body fat reduction upon C4R1 treatment was associated with improved lipid utilization estimated from decrease in respiratory quotient to the minimal level (0.7). Interestingly, the classical uncoupler 2,4-dinitrophenol at similar dose (27 µmol/kg daily) did not have any effect. Our results are relevant to the search for substances causing mild uncoupling of mitochondria that could be a promising therapeutic strategy to treat obesity.
Collapse
Affiliation(s)
- A V Kalinovich
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm, Sweden.
| | | |
Collapse
|
408
|
Min SY, Kady J, Nam M, Rojas-Rodriguez R, Berkenwald A, Kim JH, Noh HL, Kim JK, Cooper MP, Fitzgibbons T, Brehm MA, Corvera S. Human 'brite/beige' adipocytes develop from capillary networks, and their implantation improves metabolic homeostasis in mice. Nat Med 2016; 22:312-8. [PMID: 26808348 PMCID: PMC4777633 DOI: 10.1038/nm.4031] [Citation(s) in RCA: 243] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/14/2015] [Indexed: 12/15/2022]
Abstract
Uncoupling protein 1 (UCP1) is highly expressed in brown adipose tissue, where it generates heat by uncoupling electron transport from ATP production. UCP1 is also found outside classical brown adipose tissue depots, in adipocytes that are termed 'brite' (brown-in-white) or 'beige'. In humans, the presence of brite or beige (brite/beige) adipocytes is correlated with a lean, metabolically healthy phenotype, but whether a causal relationship exists is not clear. Here we report that human brite/beige adipocyte progenitors proliferate in response to pro-angiogenic factors, in association with expanding capillary networks. Adipocytes formed from these progenitors transform in response to adenylate cyclase activation from being UCP1 negative to being UCP1 positive, which is a defining feature of the beige/brite phenotype, while displaying uncoupled respiration. When implanted into normal chow-fed, or into high-fat diet (HFD)-fed, glucose-intolerant NOD-scid IL2rg(null) (NSG) mice, brite/beige adipocytes activated in vitro enhance systemic glucose tolerance. These adipocytes express neuroendocrine and secreted factors, including the pro-protein convertase PCSK1, which is strongly associated with human obesity. Pro-angiogenic conditions therefore drive the proliferation of human beige/brite adipocyte progenitors, and activated beige/brite adipocytes can affect systemic glucose homeostasis, potentially through a neuroendocrine mechanism.
Collapse
Affiliation(s)
- So Yun Min
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
- Graduate School of Biomedical Sciences, University of Massachusetts Medical School, Worcester, MA
| | - Jamie Kady
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
- Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA
| | - Minwoo Nam
- Graduate School of Biomedical Sciences, University of Massachusetts Medical School, Worcester, MA
- Cardiovascular Center of Excellence, University of Massachusetts Medical School, Worcester, MA
| | - Raziel Rojas-Rodriguez
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
- Graduate School of Biomedical Sciences, University of Massachusetts Medical School, Worcester, MA
| | - Aaron Berkenwald
- Clinical Translational Research Pathway, University of Massachusetts Medical School, Worcester, MA
| | - Jong Hun Kim
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Hye-Lim Noh
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Jason K. Kim
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Marcus P. Cooper
- Cardiovascular Center of Excellence, University of Massachusetts Medical School, Worcester, MA
| | - Timothy Fitzgibbons
- Cardiovascular Center of Excellence, University of Massachusetts Medical School, Worcester, MA
| | - Michael A. Brehm
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
- Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA
| | - Silvia Corvera
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
409
|
Turner JB, Kumar A, Koch CA. The effects of indoor and outdoor temperature on metabolic rate and adipose tissue - the Mississippi perspective on the obesity epidemic. Rev Endocr Metab Disord 2016; 17:61-71. [PMID: 27165258 DOI: 10.1007/s11154-016-9358-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Global warming, primarily caused by emissions of too much carbon dioxide, and climate change is a reality. This will lead to more extreme weather events with heatwaves and flooding. Some studies propose an association between thermal exposures and the prevalence of obesity with an increasing trend towards time spent in the thermal comfort zone. Longterm exposure to the thermal comfort zone can lead to a reduction of brown adipose tissue activity with an impact on energy expenditure and thermogenesis. Reduced seasonal cold exposure in combination with reduced diet-induced thermogenesis by a highly palatable high-fat and high-sugar diet and reduced physical activity contribute to the prevalence of obesity and the metabolic syndrome.
Collapse
Affiliation(s)
- J B Turner
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - A Kumar
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Division of Endocrinology, Diabetes, and Metabolism, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS, 39216, USA
| | - C A Koch
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA.
- Division of Endocrinology, Diabetes, and Metabolism, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
410
|
Zhao S, Mugabo Y, Ballentine G, Attane C, Iglesias J, Poursharifi P, Zhang D, Nguyen T, Erb H, Prentki R, Peyot ML, Joly E, Tobin S, Fulton S, Brown J, Madiraju S, Prentki M. α/β-Hydrolase Domain 6 Deletion Induces Adipose Browning and Prevents Obesity and Type 2 Diabetes. Cell Rep 2016; 14:2872-88. [DOI: 10.1016/j.celrep.2016.02.076] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 12/21/2015] [Accepted: 02/18/2016] [Indexed: 01/22/2023] Open
|
411
|
Morita S, Nakabayashi K, Kawai T, Hayashi K, Horii T, Kimura M, Kamei Y, Ogawa Y, Hata K, Hatada I. Gene expression profiling of white adipose tissue reveals paternal transmission of proneness to obesity. Sci Rep 2016; 6:21693. [PMID: 26868178 PMCID: PMC4751506 DOI: 10.1038/srep21693] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/29/2016] [Indexed: 11/21/2022] Open
Abstract
Previously, we found that C57BL/6J (B6) mice are more prone to develop obesity than PWK mice. In addition, we analyzed reciprocal crosses between these mice and found that (PWK × B6) F1 mice, which have B6 fathers, are more likely to develop dietary obesity than (B6 × PWK) F1 mice, which have B6 mothers. These results suggested that diet-induced obesity is paternally transmitted. In this study, we performed transcriptome analysis of adipose tissues of B6, PWK, (PWK × B6) F1, and (B6 × PWK) F1 mice using next-generation sequencing. We found that paternal transmission of diet-induced obesity was correlated with genes involved in adipose tissue inflammation, metal ion transport, and cilia. Furthermore, we analyzed the imprinted genes expressed in white adipose tissue (WAT) and obesity. Expression of paternally expressed imprinted genes (PEGs) was negatively correlated with body weight, whereas expression of maternally expressed imprinted genes (MEGs) was positively correlated. In the obesity-prone B6 mice, expression of PEGs was down-regulated by a high-fat diet, suggesting that abnormally low expression of PEGs contributes to high-fat diet-induced obesity in B6 mice. In addition, using single-nucleotide polymorphisms that differ between B6 and PWK, we identified candidate imprinted genes in WAT.
Collapse
Affiliation(s)
- Sumiyo Morita
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi Maebashi, 371-8512, Japan
| | - Kazuhiko Nakabayashi
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, 2-10-1 Okura Setagaya-ku Tokyo, 157-8535, Japan
| | - Tomoko Kawai
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, 2-10-1 Okura Setagaya-ku Tokyo, 157-8535, Japan
| | - Keiko Hayashi
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, 2-10-1 Okura Setagaya-ku Tokyo, 157-8535, Japan
| | - Takuro Horii
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi Maebashi, 371-8512, Japan
| | - Mika Kimura
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi Maebashi, 371-8512, Japan
| | - Yasutomi Kamei
- Laboratory of Molecular Nutrition, Graduate School of Environmental and Life Science, Kyoto Prefectural University, 1-5 Hangi-cho, Shimogamo, Sakyo-ku, Kyoto, 606-8522, Japan
| | - Yoshihiro Ogawa
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Bunkyo-ku, Yushima, Tokyo, 113-8510, Japan
| | - Kenichiro Hata
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, 2-10-1 Okura Setagaya-ku Tokyo, 157-8535, Japan
| | - Izuho Hatada
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi Maebashi, 371-8512, Japan
| |
Collapse
|
412
|
Alvarez-Crespo M, Csikasz RI, Martínez-Sánchez N, Diéguez C, Cannon B, Nedergaard J, López M. Essential role of UCP1 modulating the central effects of thyroid hormones on energy balance. Mol Metab 2016; 5:271-282. [PMID: 27069867 PMCID: PMC4812006 DOI: 10.1016/j.molmet.2016.01.008] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 01/18/2016] [Accepted: 01/22/2016] [Indexed: 01/05/2023] Open
Abstract
Objective Classically, metabolic effects of thyroid hormones (THs) have been considered to be peripherally mediated, i.e. different tissues in the body respond directly to thyroid hormones with an increased metabolism. An alternative view is that the metabolic effects are centrally regulated. We have examined here the degree to which prolonged, centrally infused triiodothyronine (T3) could in itself induce total body metabolic effects and the degree to which brown adipose tissue (BAT) thermogenesis was essential for such effects, by examining uncoupling protein 1 (UCP1) KO mice. Methods Wildtype and UPC1 KO mice were centrally-treated with T3 by using minipumps. Metabolic measurements were analyzed by indirect calorimetry and expression analysis by RT-PCR or western blot. BAT morphology and histology were studied by immunohistochemistry. Results We found that central T3-treatment led to reduced levels of hypothalamic AMP-activated protein kinase (AMPK) and elevated body temperature (0.7 °C). UCP1 was essential for the T3-induced increased rate of energy expenditure, which was only observable at thermoneutrality and notably only during the active phase, for the increased body weight loss, for the increased hypothalamic levels of neuropeptide Y (NPY) and agouti-related peptide (AgRP) and for the increased food intake induced by central T3-treatment. Prolonged central T3-treatment also led to recruitment of BAT and britening/beiging (“browning”) of inguinal white adipose tissue (iWAT). Conclusions We conclude that UCP1 is essential for mediation of the central effects of thyroid hormones on energy balance, and we suggest that similar UCP1-dependent effects may underlie central energy balance effects of other agents. Central thyroid hormone (TH) increases brown adipose tissue (BAT) thermogenesis. Central TH increases oxygen consumption and energy expenditure. Central TH increases food intake. Central TH promotes britening/beiging of white adipose tissue (WAT). All these metabolic effects of central TH are lost in UCP1 knockout mice.
Collapse
Affiliation(s)
- Mayte Alvarez-Crespo
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Robert I Csikasz
- The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, SE-106 91, Stockholm, Sweden
| | - Noelia Martínez-Sánchez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Barbara Cannon
- The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, SE-106 91, Stockholm, Sweden
| | - Jan Nedergaard
- The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, SE-106 91, Stockholm, Sweden.
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain.
| |
Collapse
|
413
|
Pisani DF, Beranger GE, Corinus A, Giroud M, Ghandour RA, Altirriba J, Chambard JC, Mazure NM, Bendahhou S, Duranton C, Michiels JF, Frontini A, Rohner-Jeanrenaud F, Cinti S, Christian M, Barhanin J, Amri EZ. The K+ channel TASK1 modulates β-adrenergic response in brown adipose tissue through the mineralocorticoid receptor pathway. FASEB J 2016; 30:909-922. [PMID: 26527067 DOI: 10.1096/fj.15-277475] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 10/19/2015] [Indexed: 01/26/2023]
Abstract
Brown adipose tissue (BAT) is essential for adaptive thermogenesis and dissipation of caloric excess through the activity of uncoupling protein (UCP)-1. BAT in humans is of great interest for the treatment of obesity and related diseases. In this study, the expression of Twik-related acid-sensitive K(+) channel (TASK)-1 [a pH-sensitive potassium channel encoded by the potassium channel, 2-pore domain, subfamily K, member 3 (Kcnk3) gene] correlated highly with Ucp1 expression in obese and cold-exposed mice. In addition, Task1-null mice, compared with their controls, became overweight, mainly because of an increase in white adipose tissue mass and BAT whitening. Task1(-/-)-mouse-derived brown adipocytes, compared with wild-type mouse-derived brown adipocytes, displayed an impaired β3-adrenergic receptor response that was characterized by a decrease in oxygen consumption, Ucp1 expression, and lipolysis. This phenotype was thought to be caused by an exacerbation of mineralocorticoid receptor (MR) signaling, given that it was mimicked by corticoids and reversed by an MR inhibitor. We concluded that the K(+) channel TASK1 controls the thermogenic activity in brown adipocytes through modulation of β-adrenergic receptor signaling.
Collapse
MESH Headings
- Adipocytes, Brown/cytology
- Adipocytes, Brown/metabolism
- Adipose Tissue, Brown/cytology
- Adipose Tissue, Brown/metabolism
- Animals
- Female
- Mice
- Mice, Knockout
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Oxygen Consumption/physiology
- Potassium Channels, Tandem Pore Domain/genetics
- Potassium Channels, Tandem Pore Domain/metabolism
- Receptors, Adrenergic, beta-3/metabolism
- Receptors, Mineralocorticoid/genetics
- Receptors, Mineralocorticoid/metabolism
- Signal Transduction/physiology
- Thermogenesis/physiology
Collapse
Affiliation(s)
- Didier F Pisani
- *University of Nice Sophia Antipolis, Nice, France; Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Valrose (iBV), Unité Mixte de Recherche (UMR) 7277, Nice, France; U1091, iBV, INSERM, Nice, France; UMR 7370 and Laboratories of Excellence, Ion Channel Science and Therapeutics, Laboratoire de PhysioMédecine Moléculaire (LP2M), CNRS, Nice, France; Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland, UMR 7284 and **U1081, CNRS, Institute for Research in Cancer and Aging in Nice, INSERM, Nice, France; Anatomopathology Service, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; Obesity Center, Department of Experimental and Clinical Medicine, Ancona, Italy; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Guillaume E Beranger
- *University of Nice Sophia Antipolis, Nice, France; Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Valrose (iBV), Unité Mixte de Recherche (UMR) 7277, Nice, France; U1091, iBV, INSERM, Nice, France; UMR 7370 and Laboratories of Excellence, Ion Channel Science and Therapeutics, Laboratoire de PhysioMédecine Moléculaire (LP2M), CNRS, Nice, France; Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland, UMR 7284 and **U1081, CNRS, Institute for Research in Cancer and Aging in Nice, INSERM, Nice, France; Anatomopathology Service, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; Obesity Center, Department of Experimental and Clinical Medicine, Ancona, Italy; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Alain Corinus
- *University of Nice Sophia Antipolis, Nice, France; Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Valrose (iBV), Unité Mixte de Recherche (UMR) 7277, Nice, France; U1091, iBV, INSERM, Nice, France; UMR 7370 and Laboratories of Excellence, Ion Channel Science and Therapeutics, Laboratoire de PhysioMédecine Moléculaire (LP2M), CNRS, Nice, France; Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland, UMR 7284 and **U1081, CNRS, Institute for Research in Cancer and Aging in Nice, INSERM, Nice, France; Anatomopathology Service, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; Obesity Center, Department of Experimental and Clinical Medicine, Ancona, Italy; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Maude Giroud
- *University of Nice Sophia Antipolis, Nice, France; Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Valrose (iBV), Unité Mixte de Recherche (UMR) 7277, Nice, France; U1091, iBV, INSERM, Nice, France; UMR 7370 and Laboratories of Excellence, Ion Channel Science and Therapeutics, Laboratoire de PhysioMédecine Moléculaire (LP2M), CNRS, Nice, France; Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland, UMR 7284 and **U1081, CNRS, Institute for Research in Cancer and Aging in Nice, INSERM, Nice, France; Anatomopathology Service, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; Obesity Center, Department of Experimental and Clinical Medicine, Ancona, Italy; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Rayane A Ghandour
- *University of Nice Sophia Antipolis, Nice, France; Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Valrose (iBV), Unité Mixte de Recherche (UMR) 7277, Nice, France; U1091, iBV, INSERM, Nice, France; UMR 7370 and Laboratories of Excellence, Ion Channel Science and Therapeutics, Laboratoire de PhysioMédecine Moléculaire (LP2M), CNRS, Nice, France; Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland, UMR 7284 and **U1081, CNRS, Institute for Research in Cancer and Aging in Nice, INSERM, Nice, France; Anatomopathology Service, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; Obesity Center, Department of Experimental and Clinical Medicine, Ancona, Italy; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Jordi Altirriba
- *University of Nice Sophia Antipolis, Nice, France; Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Valrose (iBV), Unité Mixte de Recherche (UMR) 7277, Nice, France; U1091, iBV, INSERM, Nice, France; UMR 7370 and Laboratories of Excellence, Ion Channel Science and Therapeutics, Laboratoire de PhysioMédecine Moléculaire (LP2M), CNRS, Nice, France; Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland, UMR 7284 and **U1081, CNRS, Institute for Research in Cancer and Aging in Nice, INSERM, Nice, France; Anatomopathology Service, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; Obesity Center, Department of Experimental and Clinical Medicine, Ancona, Italy; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Jean-Claude Chambard
- *University of Nice Sophia Antipolis, Nice, France; Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Valrose (iBV), Unité Mixte de Recherche (UMR) 7277, Nice, France; U1091, iBV, INSERM, Nice, France; UMR 7370 and Laboratories of Excellence, Ion Channel Science and Therapeutics, Laboratoire de PhysioMédecine Moléculaire (LP2M), CNRS, Nice, France; Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland, UMR 7284 and **U1081, CNRS, Institute for Research in Cancer and Aging in Nice, INSERM, Nice, France; Anatomopathology Service, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; Obesity Center, Department of Experimental and Clinical Medicine, Ancona, Italy; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Nathalie M Mazure
- *University of Nice Sophia Antipolis, Nice, France; Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Valrose (iBV), Unité Mixte de Recherche (UMR) 7277, Nice, France; U1091, iBV, INSERM, Nice, France; UMR 7370 and Laboratories of Excellence, Ion Channel Science and Therapeutics, Laboratoire de PhysioMédecine Moléculaire (LP2M), CNRS, Nice, France; Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland, UMR 7284 and **U1081, CNRS, Institute for Research in Cancer and Aging in Nice, INSERM, Nice, France; Anatomopathology Service, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; Obesity Center, Department of Experimental and Clinical Medicine, Ancona, Italy; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Saïd Bendahhou
- *University of Nice Sophia Antipolis, Nice, France; Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Valrose (iBV), Unité Mixte de Recherche (UMR) 7277, Nice, France; U1091, iBV, INSERM, Nice, France; UMR 7370 and Laboratories of Excellence, Ion Channel Science and Therapeutics, Laboratoire de PhysioMédecine Moléculaire (LP2M), CNRS, Nice, France; Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland, UMR 7284 and **U1081, CNRS, Institute for Research in Cancer and Aging in Nice, INSERM, Nice, France; Anatomopathology Service, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; Obesity Center, Department of Experimental and Clinical Medicine, Ancona, Italy; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Christophe Duranton
- *University of Nice Sophia Antipolis, Nice, France; Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Valrose (iBV), Unité Mixte de Recherche (UMR) 7277, Nice, France; U1091, iBV, INSERM, Nice, France; UMR 7370 and Laboratories of Excellence, Ion Channel Science and Therapeutics, Laboratoire de PhysioMédecine Moléculaire (LP2M), CNRS, Nice, France; Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland, UMR 7284 and **U1081, CNRS, Institute for Research in Cancer and Aging in Nice, INSERM, Nice, France; Anatomopathology Service, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; Obesity Center, Department of Experimental and Clinical Medicine, Ancona, Italy; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Jean-François Michiels
- *University of Nice Sophia Antipolis, Nice, France; Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Valrose (iBV), Unité Mixte de Recherche (UMR) 7277, Nice, France; U1091, iBV, INSERM, Nice, France; UMR 7370 and Laboratories of Excellence, Ion Channel Science and Therapeutics, Laboratoire de PhysioMédecine Moléculaire (LP2M), CNRS, Nice, France; Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland, UMR 7284 and **U1081, CNRS, Institute for Research in Cancer and Aging in Nice, INSERM, Nice, France; Anatomopathology Service, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; Obesity Center, Department of Experimental and Clinical Medicine, Ancona, Italy; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Andrea Frontini
- *University of Nice Sophia Antipolis, Nice, France; Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Valrose (iBV), Unité Mixte de Recherche (UMR) 7277, Nice, France; U1091, iBV, INSERM, Nice, France; UMR 7370 and Laboratories of Excellence, Ion Channel Science and Therapeutics, Laboratoire de PhysioMédecine Moléculaire (LP2M), CNRS, Nice, France; Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland, UMR 7284 and **U1081, CNRS, Institute for Research in Cancer and Aging in Nice, INSERM, Nice, France; Anatomopathology Service, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; Obesity Center, Department of Experimental and Clinical Medicine, Ancona, Italy; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Françoise Rohner-Jeanrenaud
- *University of Nice Sophia Antipolis, Nice, France; Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Valrose (iBV), Unité Mixte de Recherche (UMR) 7277, Nice, France; U1091, iBV, INSERM, Nice, France; UMR 7370 and Laboratories of Excellence, Ion Channel Science and Therapeutics, Laboratoire de PhysioMédecine Moléculaire (LP2M), CNRS, Nice, France; Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland, UMR 7284 and **U1081, CNRS, Institute for Research in Cancer and Aging in Nice, INSERM, Nice, France; Anatomopathology Service, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; Obesity Center, Department of Experimental and Clinical Medicine, Ancona, Italy; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Saverio Cinti
- *University of Nice Sophia Antipolis, Nice, France; Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Valrose (iBV), Unité Mixte de Recherche (UMR) 7277, Nice, France; U1091, iBV, INSERM, Nice, France; UMR 7370 and Laboratories of Excellence, Ion Channel Science and Therapeutics, Laboratoire de PhysioMédecine Moléculaire (LP2M), CNRS, Nice, France; Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland, UMR 7284 and **U1081, CNRS, Institute for Research in Cancer and Aging in Nice, INSERM, Nice, France; Anatomopathology Service, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; Obesity Center, Department of Experimental and Clinical Medicine, Ancona, Italy; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Mark Christian
- *University of Nice Sophia Antipolis, Nice, France; Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Valrose (iBV), Unité Mixte de Recherche (UMR) 7277, Nice, France; U1091, iBV, INSERM, Nice, France; UMR 7370 and Laboratories of Excellence, Ion Channel Science and Therapeutics, Laboratoire de PhysioMédecine Moléculaire (LP2M), CNRS, Nice, France; Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland, UMR 7284 and **U1081, CNRS, Institute for Research in Cancer and Aging in Nice, INSERM, Nice, France; Anatomopathology Service, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; Obesity Center, Department of Experimental and Clinical Medicine, Ancona, Italy; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Jacques Barhanin
- *University of Nice Sophia Antipolis, Nice, France; Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Valrose (iBV), Unité Mixte de Recherche (UMR) 7277, Nice, France; U1091, iBV, INSERM, Nice, France; UMR 7370 and Laboratories of Excellence, Ion Channel Science and Therapeutics, Laboratoire de PhysioMédecine Moléculaire (LP2M), CNRS, Nice, France; Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland, UMR 7284 and **U1081, CNRS, Institute for Research in Cancer and Aging in Nice, INSERM, Nice, France; Anatomopathology Service, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; Obesity Center, Department of Experimental and Clinical Medicine, Ancona, Italy; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Ez-Zoubir Amri
- *University of Nice Sophia Antipolis, Nice, France; Centre National de la Recherche Scientifique (CNRS), Institut de Biologie Valrose (iBV), Unité Mixte de Recherche (UMR) 7277, Nice, France; U1091, iBV, INSERM, Nice, France; UMR 7370 and Laboratories of Excellence, Ion Channel Science and Therapeutics, Laboratoire de PhysioMédecine Moléculaire (LP2M), CNRS, Nice, France; Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland, UMR 7284 and **U1081, CNRS, Institute for Research in Cancer and Aging in Nice, INSERM, Nice, France; Anatomopathology Service, Pasteur Hospital, Centre Hospitalier Universitaire de Nice, Nice, France; Obesity Center, Department of Experimental and Clinical Medicine, Ancona, Italy; Warwick Medical School, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
414
|
Chabowska‐Kita A, Kozak LP. The critical period for brown adipocyte development: Genetic and environmental influences. Obesity (Silver Spring) 2016; 24:283-90. [PMID: 26813522 PMCID: PMC4744992 DOI: 10.1002/oby.21376] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 09/27/2015] [Indexed: 12/30/2022]
Abstract
OBJECTIVE The current review summarizes recent advances in the origin of brown adipocytes in rodents and humans. METHODS This review describes recent insights into induction of the brown adipocyte phenotype (BAP) in white fat (WAT) revealed by murine studies during the early postnatal period and reversible temperature transitions. The origin of adipocytes and identity of progenitors as indicated by lineage tracing experiments are reviewed. RESULTS We describe a genetic model for brown adipocyte development that involves the appearance of brown adipocytes in WAT at 21 days of age and a mechanism of post-weaning involution relevant for acquisition of the BAP in fully functional WAT in mice. Under normal physiological conditions, the BAP is dormant with the potential to be stimulated by changes in the external environment. Current evidence for the acquisition of brown adipocytes by interconversion of mature adipocytes versus de novo recruitment of progenitors suggests that mechanisms for acquisition of the BAP in WAT in mice are depot-specific and controlled by allelic variation. CONCLUSIONS Although the BAP is highly variable among mice, there is no information on genetic variability in the expression of brown adipocytes in humans. Thus, deeper understanding of genetic mechanisms underlying development of functional brown adipocytes is crucial.
Collapse
Affiliation(s)
| | - Leslie P. Kozak
- Institute of Animal Reproduction and Food Research, Polish Academy of SciencesOlsztynPoland
| |
Collapse
|
415
|
Barbagallo I, Vanella L, Cambria MT, Tibullo D, Godos J, Guarnaccia L, Zappalà A, Galvano F, Li Volti G. Silibinin Regulates Lipid Metabolism and Differentiation in Functional Human Adipocytes. Front Pharmacol 2016; 6:309. [PMID: 26834634 PMCID: PMC4720740 DOI: 10.3389/fphar.2015.00309] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 12/14/2015] [Indexed: 12/21/2022] Open
Abstract
Silibinin, a natural plant flavonolignan is the main active constituent found in milk thistle (Silybum marianum). It is known to have hepatoprotective, anti-neoplastic effect, and suppresses lipid accumulation in adipocytes. Objective of this study was to investigate the effect of silibinin on adipogenic differentiation and thermogenic capacity of human adipose tissue derived mesenchymal stem cells. Silibinin (10 μM) treatment, either at the beginning or at the end of adipogenic differentiation, resulted in an increase of SIRT-1, PPARα, Pgc-1α, and UCPs gene expression. Moreover, silibinin administration resulted in a decrease of PPARγ, FABP4, FAS, and MEST/PEG1 gene expression during the differentiation, confirming that this compound is able to reduce fatty acid accumulation and adipocyte size. Our data showed that silibinin regulated adipocyte lipid metabolism, inducing thermogenesis and promoting a brown remodeling in adipocyte. Taken together, our findings suggest that silibinin increases UCPs expression by stimulation of SIRT1, PPARα, and Pgc-1α, improved metabolic parameters, decreased lipid mass leading to the formation of functional adipocytes.
Collapse
Affiliation(s)
- Ignazio Barbagallo
- Biochemistry Section, Department of Drug Science, University of CataniaCatania, Italy
| | - Luca Vanella
- Biochemistry Section, Department of Drug Science, University of CataniaCatania, Italy
| | - Maria T. Cambria
- Department of Biomedical and Biotechnological Sciences, University of CataniaCatania, Italy
| | | | - Justyna Godos
- Department of Biomedical and Biotechnological Sciences, University of CataniaCatania, Italy
| | - Laura Guarnaccia
- Department of Biomedical and Biotechnological Sciences, University of CataniaCatania, Italy
| | - Agata Zappalà
- Department of Biomedical and Biotechnological Sciences, University of CataniaCatania, Italy
| | - Fabio Galvano
- Department of Biomedical and Biotechnological Sciences, University of CataniaCatania, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of CataniaCatania, Italy
| |
Collapse
|
416
|
Krott LM, Piscitelli F, Heine M, Borrino S, Scheja L, Silvestri C, Heeren J, Di Marzo V. Endocannabinoid regulation in white and brown adipose tissue following thermogenic activation. J Lipid Res 2016; 57:464-73. [PMID: 26768656 DOI: 10.1194/jlr.m065227] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Indexed: 12/17/2022] Open
Abstract
The endocannabinoids and their main receptor, cannabinoid type-1 (CB1), suppress intracellular cyclic AMP levels and have emerged as key players in the control of energy metabolism. CB1 agonists and blockers have been reported to influence the thermogenic function of white and brown adipose tissue (WAT and BAT), affecting body weight through the inhibition and stimulation of energy expenditure, respectively. The purpose of the current study was to investigate the regulation of the endocannabinoid system in WAT and BAT following exposure to either cold or specific agonism of β3-adrenoceptors using CL316,243 (CL), conditions known to cause BAT activation and WAT browning. To address this question, we performed quantitative PCR-based mRNA profiling of genes important for endocannabinoid synthesis, degradation, and signaling, and determined endocannabinoid levels by LC-MS in WAT and BAT of control, cold-exposed, and CL-treated wild-type mice as well as primary brown adipocytes. Treatment with CL and exposure to cold caused an upregulation of endocannabinoid levels and biosynthetic enzymes in WAT. Acute β3-adrenoceptor activation increased endocannabinoids and a subset of genes of biosynthesis in BAT and primary brown adipocytes. We suggest that the cold-mediated increase in endocannabinoid tone is part of autocrine negative feed-back mechanisms controlling β3-adrenoceptor-induced BAT activation and WAT browning.
Collapse
Affiliation(s)
- Lucia M Krott
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany
| | - Fabiana Piscitelli
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, 80078 Pozzuoli, Naples, Italy
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany
| | - Simona Borrino
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, 80078 Pozzuoli, Naples, Italy
| | - Ludger Scheja
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany
| | - Cristoforo Silvestri
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, 80078 Pozzuoli, Naples, Italy
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg Eppendorf, 20246 Hamburg, Germany
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, 80078 Pozzuoli, Naples, Italy
| |
Collapse
|
417
|
Lactate induces FGF21 expression in adipocytes through a p38-MAPK pathway. Biochem J 2016; 473:685-92. [PMID: 26769382 DOI: 10.1042/bj20150808] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 01/14/2016] [Indexed: 01/22/2023]
Abstract
FGF21 (fibroblast growth factor 21), first described as a main fasting-responsive molecule in the liver, has been shown to act as a true metabolic regulator in additional tissues, including muscle and adipose tissues. In the present study, we found that the expression and secretion of FGF21 was very rapidly increased following lactate exposure in adipocytes. Using different pharmacological and knockout mice models, we demonstrated that lactate regulates Fgf21 expression through a NADH/NAD-independent pathway, but requires active p38-MAPK (mitogen activated protein kinase) signalling. We also demonstrated that this effect is not restricted to lactate as additional metabolites including pyruvate and ketone bodies also activated the FGF21 stress response. FGF21 release by adipose cells in response to an excess of intermediate metabolites may represent a physiological mechanism by which the sensing of environmental metabolic conditions results in the release of FGF21 to improve metabolic adaptations.
Collapse
|
418
|
Ghandour RA, Giroud M, Vegiopoulos A, Herzig S, Ailhaud G, Amri EZ, Pisani DF. IP-receptor and PPARs trigger the conversion of human white to brite adipocyte induced by carbaprostacyclin. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:285-93. [PMID: 26775637 DOI: 10.1016/j.bbalip.2016.01.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 01/04/2016] [Accepted: 01/12/2016] [Indexed: 12/31/2022]
Abstract
Brite adipocytes recently discovered in humans are of considerable importance in energy expenditure by converting energy excess into heat. This property could be useful in the treatment of obesity, and nutritional aspects are relevant to this important issue. Using hMADS cells as a human cell model which undergoes a white to a brite adipocyte conversion, we had shown previously that arachidonic acid, the major metabolite of the essential nutrient Ω6-linoleic acid, plays a major role in this process. Its metabolites PGE2 and PGF2 alpha inhibit this process via a calcium-dependent pathway, whereas in contrast carbaprostacyclin (cPGI2), a stable analog of prostacyclin, activates white to brite adipocyte conversion. Herein, we show that cPGI2 generates via its cognate cell-surface receptor IP-R, a cyclic AMP-signaling pathway involving PKA activity which in turn induces the expression of UCP1. In addition, cPGI2 activates the pathway of nuclear receptors of the PPAR family, i.e. PPARα and PPARγ, which act separately from IP-R to up-regulate the expression of key genes involved in the function of brite adipocytes. Thus dual pathways are playing in concert for the occurrence of a browning process of human white adipocytes. These results make prostacyclin analogs as a new class of interesting molecules to treat obesity and associated diseases.
Collapse
Affiliation(s)
- Rayane A Ghandour
- Univ. Nice Sophia Antipolis, iBV, UMR 7277, Nice, France; CNRS, iBV UMR 7277, Nice, France; Inserm, iBV, U1091, Nice, France
| | - Maude Giroud
- Univ. Nice Sophia Antipolis, iBV, UMR 7277, Nice, France; CNRS, iBV UMR 7277, Nice, France; Inserm, iBV, U1091, Nice, France
| | - Alexandros Vegiopoulos
- DKFZ Junior Group Metabolism and Stem Cell Plasticity, German Cancer Research Center, Heidelberg, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany; Molecular Metabolic Control, Medical Faculty, Technical University Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Gérard Ailhaud
- Univ. Nice Sophia Antipolis, iBV, UMR 7277, Nice, France; CNRS, iBV UMR 7277, Nice, France; Inserm, iBV, U1091, Nice, France
| | - Ez-Zoubir Amri
- Univ. Nice Sophia Antipolis, iBV, UMR 7277, Nice, France; CNRS, iBV UMR 7277, Nice, France; Inserm, iBV, U1091, Nice, France.
| | - Didier F Pisani
- Univ. Nice Sophia Antipolis, iBV, UMR 7277, Nice, France; CNRS, iBV UMR 7277, Nice, France; Inserm, iBV, U1091, Nice, France.
| |
Collapse
|
419
|
Mouse strains to study cold-inducible beige progenitors and beige adipocyte formation and function. Nat Commun 2016; 7:10184. [PMID: 26729601 PMCID: PMC4728429 DOI: 10.1038/ncomms10184] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 11/12/2015] [Indexed: 12/29/2022] Open
Abstract
Cold temperatures induce formation of beige adipocytes, which convert glucose and fatty acids to heat, and may increase energy expenditure, reduce adiposity and lower blood glucose. This therapeutic potential is unrealized, hindered by a dearth of genetic tools to fate map, track and manipulate beige progenitors and ‘beiging'. Here we examined 12 Cre/inducible Cre mouse strains that mark adipocyte, muscle and mural lineages, three proposed beige origins. Among these mouse strains, only those that marked perivascular mural cells tracked the cold-induced beige lineage. Two SMA-based strains, SMA-CreERT2 and SMA-rtTA, fate mapped into the majority of cold-induced beige adipocytes and SMA-marked progenitors appeared essential for beiging. Disruption of the potential of the SMA-tracked progenitors to form beige adipocytes was accompanied by an inability to maintain body temperature and by hyperglycaemia. Thus, SMA-engineered mice may be useful to track and manipulate beige progenitors, beige adipocyte formation and function. Beige adipocytes are formed in response to cold and thought to contribute to organismal energy homeostasis. Here, the authors study a range of conditional and inducible RFP-expressing Cre mouse strains and find that SMA-based lines are the most useful for mapping beige adipocyte progenitor cells.
Collapse
|
420
|
Hilse KE, Kalinovich AV, Rupprecht A, Smorodchenko A, Zeitz U, Staniek K, Erben RG, Pohl EE. The expression of UCP3 directly correlates to UCP1 abundance in brown adipose tissue. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1857:72-78. [PMID: 26518386 PMCID: PMC7115856 DOI: 10.1016/j.bbabio.2015.10.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/16/2015] [Accepted: 10/25/2015] [Indexed: 01/14/2023]
Abstract
UCP1 and UCP3 are members of the uncoupling protein (UCP) subfamily and are localized in the inner mitochondrial membrane. Whereas UCP1's central role in non-shivering thermogenesis is acknowledged, the function and even tissue expression pattern of UCP3 are still under dispute. Because UCP3 properties regarding transport of protons are qualitatively identical to those of UCP1, its expression in brown adipose tissue (BAT) alongside UCP1 requires justification. In this work, we tested whether any correlation exists between the expression of UCP1 and UCP3 in BAT by quantification of protein amounts in mouse tissues at physiological conditions, in cold-acclimated and UCP1 knockout mice. Quantification using recombinant UCP3 revealed that the UCP3 amount in BAT (0.51ng/(μg total tissue protein)) was nearly one order of magnitude higher than that in muscles and heart. Cold-acclimated mice showed an approximate three-fold increase in UCP3 abundance in BAT in comparison to mice in thermoneutral conditions. Surprisingly, we found a significant decrease of UCP3 in BAT of UCP1 knockout mice, whereas the protein amount in skeletal and heart muscles remained constant. UCP3 abundance decreased even more in cold-acclimated UCP1 knockout mice. Protein quantification in UCP3 knockout mice revealed no compensatory increase in UCP1 or UCP2 expression. Our results do not support the participation of UCP3 in thermogenesis in the absence of UCP1 in BAT, but clearly demonstrate the correlation in abundance between both proteins. The latter is important for understanding UCP3's function in BAT.
Collapse
Affiliation(s)
- Karolina E Hilse
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Anastasia V Kalinovich
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Anne Rupprecht
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Alina Smorodchenko
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Ute Zeitz
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Katrin Staniek
- Institute of Pharmacology and Toxicology, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Reinhold G Erben
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Elena E Pohl
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria.
| |
Collapse
|
421
|
Fish oil intake induces UCP1 upregulation in brown and white adipose tissue via the sympathetic nervous system. Sci Rep 2015; 5:18013. [PMID: 26673120 PMCID: PMC4682086 DOI: 10.1038/srep18013] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 11/10/2015] [Indexed: 12/11/2022] Open
Abstract
Brown adipose tissue (BAT) plays a central role in regulating energy homeostasis, and may provide novel strategies for the treatment of human obesity. BAT-mediated thermogenesis is regulated by mitochondrial uncoupling protein 1 (UCP1) in classical brown and ectopic beige adipocytes, and is controlled by sympathetic nervous system (SNS). Previous work indicated that fish oil intake reduces fat accumulation and induces UCP1 expression in BAT; however, the detailed mechanism of this effect remains unclear. In this study, we investigated the effect of fish oil on energy expenditure and the SNS. Fish oil intake increased oxygen consumption and rectal temperature, with concomitant upregulation of UCP1 and the β3 adrenergic receptor (β3AR), two markers of beige adipocytes, in the interscapular BAT and inguinal white adipose tissue (WAT). Additionally, fish oil intake increased the elimination of urinary catecholamines and the noradrenaline (NA) turnover rate in interscapular BAT and inguinal WAT. Furthermore, the effects of fish oil on SNS-mediated energy expenditure were abolished in transient receptor potential vanilloid 1 (TRPV1) knockout mice. In conclusion, fish oil intake can induce UCP1 expression in classical brown and beige adipocytes via the SNS, thereby attenuating fat accumulation and ameliorating lipid metabolism.
Collapse
|
422
|
Divergent responses to thermogenic stimuli in BAT and subcutaneous adipose tissue from interleukin 18 and interleukin 18 receptor 1-deficient mice. Sci Rep 2015; 5:17977. [PMID: 26656097 PMCID: PMC4674707 DOI: 10.1038/srep17977] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 11/10/2015] [Indexed: 01/15/2023] Open
Abstract
Brown and beige adipocytes recruitment in brown (BAT) or white adipose tissue, mainly in the inguinal fat pad (iWAT), meet the need for temperature adaptation in cold-exposure conditions and protect against obesity in face of hypercaloric diets. Using interleukin18 (Il18) and Il18 receptor 1- knockout (Il18r1-KO) mice, this study aimed to investigate the role of IL18 signaling in BAT and iWAT activation and thermogenesis under both stimuli. Il18-KO, extremely dietary obesity-prone as previously described, failed to develop diet-induced thermogenesis as assessed by BAT and iWAT Ucp1 mRNA levels. Overweight when fed standard chow but not HFD, HFD-fed Il18r1-KO mice exhibited increased iWAT Ucp1 gene expression. Energy expenditure was reduced in pre-obese Il18r1-KO mice and restored upon HFD-challenge. Cold exposure lead to similar results; Il18r1-KO mice were protected against acute body temperature drop, displaying a more brown-like structure, alternative macrophage activation and thermogenic gene expression in iWAT than WT controls. Opposite effects were observed in Il18-KO mice. Thus, Il18 and Il18r1 genetic ablation disparate effects on energy homeostasis are likely mediated by divergent BAT responses to thermogenic stimuli as well as iWAT browning. These results suggest that a more complex receptor-signaling system mediates the IL18 adipose-tissue specific effects in energy expenditure.
Collapse
|
423
|
Maurer SF, Fromme T, Grossman LI, Hüttemann M, Klingenspor M. The brown and brite adipocyte marker Cox7a1 is not required for non-shivering thermogenesis in mice. Sci Rep 2015; 5:17704. [PMID: 26635001 PMCID: PMC4669493 DOI: 10.1038/srep17704] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 11/03/2015] [Indexed: 12/11/2022] Open
Abstract
The cytochrome c oxidase subunit isoform Cox7a1 is highly abundant in skeletal muscle and heart and influences enzyme activity in these tissues characterised by high oxidative capacity. We identified Cox7a1, well-known as brown adipocyte marker gene, as a cold-responsive protein of brown adipose tissue. We hypothesised a mechanistic relationship between cytochrome c oxidase activity and Cox7a1 protein levels affecting the oxidative capacity of brown adipose tissue and thus non-shivering thermogenesis. We subjected wildtype and Cox7a1 knockout mice to different temperature regimens and tested characteristics of brown adipose tissue activation. Cytochrome c oxidase activity, uncoupling protein 1 expression and maximal norepinephrine-induced heat production were gradually increased during cold-acclimation, but unaffected by Cox7a1 knockout. Moreover, the abundance of uncoupling protein 1 competent brite cells in white adipose tissue was not influenced by presence or absence of Cox7a1. Skin temperature in the interscapular region of neonates was lower in uncoupling protein 1 knockout pups employed as a positive control, but not in Cox7a1 knockout pups. Body mass gain and glucose tolerance did not differ between wildtype and Cox7a1 knockout mice fed with high fat or control diet. We conclude that brown adipose tissue function in mice does not require the presence of Cox7a1.
Collapse
Affiliation(s)
- Stefanie F Maurer
- Chair of Molecular Nutritional Medicine, Technische Universität München, Else Kröner-Fresenius Center for Nutritional Medicine &ZIEL-Institute for Food and Health, 85350 Freising-Weihenstephan, Germany
| | - Tobias Fromme
- Chair of Molecular Nutritional Medicine, Technische Universität München, Else Kröner-Fresenius Center for Nutritional Medicine &ZIEL-Institute for Food and Health, 85350 Freising-Weihenstephan, Germany
| | - Lawrence I Grossman
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Martin Klingenspor
- Chair of Molecular Nutritional Medicine, Technische Universität München, Else Kröner-Fresenius Center for Nutritional Medicine &ZIEL-Institute for Food and Health, 85350 Freising-Weihenstephan, Germany
| |
Collapse
|
424
|
Skeletal muscle mitochondrial uncoupling prevents diabetes but not obesity in NZO mice, a model for polygenic diabesity. GENES AND NUTRITION 2015; 10:57. [PMID: 26584809 DOI: 10.1007/s12263-015-0507-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/04/2015] [Indexed: 01/04/2023]
Abstract
Induction of skeletal muscle (SM) mitochondrial stress by expression of uncoupling protein 1 (UCP1) in mice results in a healthy metabolic phenotype associated with increased secretion of FGF21 from SM. Here, we investigated whether SM mitochondrial uncoupling can compensate obesity and insulin resistance in the NZO mouse, a polygenic diabesity model. Male NZO mice were crossed with heterozygous UCP1 transgenic (tg) mice (mixed C57BL/6/CBA background) and further backcrossed to obtain F1 and N2 offspring with 50 and 75 % NZO background, respectively. Male F1 and N2 progeny were fed a high-fat diet ad libitum for 20 weeks from weaning. Blood glucose was reduced, and diabetes (severe hyperglycemia >300 mg/dl) was fully prevented in both F1- and N2-tg progeny compared to a diabetes prevalence of 15 % in F1 and 42 % in N2 wild type. In contrast, relative body fat content and plasma insulin were decreased, and glucose tolerance was improved, in F1-tg only. Both F1 and N2-tg showed decreased lean body mass. Accordingly, induction of SM stress response including FGF21 expression and secretion was similar in both F1 and N2-tg mice. In white adipose tissue, expression of FGF21 target genes was enhanced in F1 and N2-tg mice, whereas lipid metabolism genes were induced in F1-tg only. There was no evidence for induction of browning in either UCP1 backcross. We conclude that SM mitochondrial uncoupling induces FGF21 expression and prevents diabetes in mice with a 50-75 % NZO background independent of its effects on adipose tissue.
Collapse
|
425
|
Patsouris D, Qi P, Abdullahi A, Stanojcic M, Chen P, Parousis A, Amini-Nik S, Jeschke MG. Burn Induces Browning of the Subcutaneous White Adipose Tissue in Mice and Humans. Cell Rep 2015; 13:1538-44. [PMID: 26586436 PMCID: PMC4662886 DOI: 10.1016/j.celrep.2015.10.028] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/09/2015] [Accepted: 10/08/2015] [Indexed: 01/22/2023] Open
Abstract
Burn is accompanied by long-lasting immunometabolic alterations referred to as hypermetabolism that are characterized by a considerable increase in resting energy expenditure and substantial whole-body catabolism. In burned patients, the length and magnitude of the hypermetabolic state is the highest of all patients and associated with profoundly increased morbidity and mortality. Unfortunately, the mechanisms involved in hypermetabolism are essentially unknown. We hypothesized that the adipose tissue plays a central role for the induction and persistence of hypermetabolism post-burn injury. Here, we show that burn induces a switch in the phenotype of the subcutaneous fat from white to beige, with associated characteristics such as increased mitochondrial mass and UCP1 expression. Our results further demonstrate the significant role of catecholamines and interleukin-6 in this process. We conclude that subcutaneous fat remodeling and browning represent an underlying mechanism that explains the elevated energy expenditure in burn-induced hypermetabolism.
Collapse
Affiliation(s)
- David Patsouris
- Department of Surgery, Division of Plastic Surgery, Department of Immunology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Peter Qi
- Department of Surgery, Division of Plastic Surgery, Department of Immunology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Abdikarim Abdullahi
- Department of Surgery, Division of Plastic Surgery, Department of Immunology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Mile Stanojcic
- Department of Surgery, Division of Plastic Surgery, Department of Immunology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Peter Chen
- Department of Surgery, Division of Plastic Surgery, Department of Immunology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Alexandra Parousis
- Department of Surgery, Division of Plastic Surgery, Department of Immunology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Saeid Amini-Nik
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; Department of Surgery, Division of Plastic Surgery, Department of Immunology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Marc G Jeschke
- Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; Department of Surgery, Division of Plastic Surgery, Department of Immunology, University of Toronto, Toronto, ON M5S 1A1, Canada.
| |
Collapse
|
426
|
van den Beukel JC, Boon MR, Steenbergen J, Rensen PCN, Meijer OC, Themmen APN, Grefhorst A. Cold Exposure Partially Corrects Disturbances in Lipid Metabolism in a Male Mouse Model of Glucocorticoid Excess. Endocrinology 2015; 156:4115-28. [PMID: 26372178 DOI: 10.1210/en.2015-1092] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
High glucocorticoid concentrations are accompanied by metabolic side effects such as high plasma triglyceride (TG) concentrations. Liver, brown adipose tissue (BAT) and white adipose tissue are important regulators of plasma TG. Exposure to 4°C reduces plasma TG concentrations, and we therefore aimed to study the interaction between glucocorticoid excess and 24 hours of exposure to 4°C on lipid metabolism. For this, mice were implanted with 50-mg corticosterone or control pellets and housed for 24 hours at 23°C or 4°C 1 week later, after which various aspects of TG metabolism in liver, BAT, and white adipose tissue were studied. Corticosterone treatment resulted in a 3.8-fold increase of plasma TG concentrations. Increased TG was normalized by cold exposure, an effect still present 24 hours after cold exposure. Corticosterone treatment increased hepatic TG content by 3.5-fold and provoked secretion of large, TG-rich very low density lipoprotein particles. Cold exposure reduced very low density lipoprotein-TG secretion by approximately 50%. Corticosterone strongly decreased BAT activity: BAT weight increased by 3.5-fold, whereas uncoupling protein 1 (Ucp1) mRNA expression and Ucp1 protein content of BAT were reduced by 75% and 60%, respectively. Cold exposure partially normalized these parameters of BAT activity. The uptake of TG by BAT was not affected by corticosterone treatment but was increased 4.5-fold upon cold exposure. In conclusion, cold exposure normalizes corticosterone-induced hypertriglyceridemia, at least partly via activating BAT.
Collapse
Affiliation(s)
- Johanna C van den Beukel
- Department of Internal Medicine (J.C.v.d.B., J.S., A.P.N.T., A.G.), Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; Department of Medicine (M.R.B., P.C.N.R., O.C.M.), Division of Endocrinology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; and Einthoven Laboratory of Experimental Vascular Medicine (M.R.B., P.C.N.R., O.C.M.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Mariëtte R Boon
- Department of Internal Medicine (J.C.v.d.B., J.S., A.P.N.T., A.G.), Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; Department of Medicine (M.R.B., P.C.N.R., O.C.M.), Division of Endocrinology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; and Einthoven Laboratory of Experimental Vascular Medicine (M.R.B., P.C.N.R., O.C.M.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Jacobie Steenbergen
- Department of Internal Medicine (J.C.v.d.B., J.S., A.P.N.T., A.G.), Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; Department of Medicine (M.R.B., P.C.N.R., O.C.M.), Division of Endocrinology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; and Einthoven Laboratory of Experimental Vascular Medicine (M.R.B., P.C.N.R., O.C.M.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Patrick C N Rensen
- Department of Internal Medicine (J.C.v.d.B., J.S., A.P.N.T., A.G.), Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; Department of Medicine (M.R.B., P.C.N.R., O.C.M.), Division of Endocrinology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; and Einthoven Laboratory of Experimental Vascular Medicine (M.R.B., P.C.N.R., O.C.M.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Onno C Meijer
- Department of Internal Medicine (J.C.v.d.B., J.S., A.P.N.T., A.G.), Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; Department of Medicine (M.R.B., P.C.N.R., O.C.M.), Division of Endocrinology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; and Einthoven Laboratory of Experimental Vascular Medicine (M.R.B., P.C.N.R., O.C.M.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Axel P N Themmen
- Department of Internal Medicine (J.C.v.d.B., J.S., A.P.N.T., A.G.), Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; Department of Medicine (M.R.B., P.C.N.R., O.C.M.), Division of Endocrinology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; and Einthoven Laboratory of Experimental Vascular Medicine (M.R.B., P.C.N.R., O.C.M.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Aldo Grefhorst
- Department of Internal Medicine (J.C.v.d.B., J.S., A.P.N.T., A.G.), Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; Department of Medicine (M.R.B., P.C.N.R., O.C.M.), Division of Endocrinology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; and Einthoven Laboratory of Experimental Vascular Medicine (M.R.B., P.C.N.R., O.C.M.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
427
|
Kazak L, Chouchani ET, Jedrychowski MP, Erickson BK, Shinoda K, Cohen P, Vetrivelan R, Lu GZ, Laznik-Bogoslavski D, Hasenfuss SC, Kajimura S, Gygi SP, Spiegelman BM. A creatine-driven substrate cycle enhances energy expenditure and thermogenesis in beige fat. Cell 2015; 163:643-55. [PMID: 26496606 DOI: 10.1016/j.cell.2015.09.035] [Citation(s) in RCA: 587] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Revised: 07/10/2015] [Accepted: 09/08/2015] [Indexed: 02/03/2023]
Abstract
Thermogenic brown and beige adipose tissues dissipate chemical energy as heat, and their thermogenic activities can combat obesity and diabetes. Herein the functional adaptations to cold of brown and beige adipose depots are examined using quantitative mitochondrial proteomics. We identify arginine/creatine metabolism as a beige adipose signature and demonstrate that creatine enhances respiration in beige-fat mitochondria when ADP is limiting. In murine beige fat, cold exposure stimulates mitochondrial creatine kinase activity and induces coordinated expression of genes associated with creatine metabolism. Pharmacological reduction of creatine levels decreases whole-body energy expenditure after administration of a β3-agonist and reduces beige and brown adipose metabolic rate. Genes of creatine metabolism are compensatorily induced when UCP1-dependent thermogenesis is ablated, and creatine reduction in Ucp1-deficient mice reduces core body temperature. These findings link a futile cycle of creatine metabolism to adipose tissue energy expenditure and thermal homeostasis. PAPERCLIP.
Collapse
Affiliation(s)
- Lawrence Kazak
- Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard University Medical School, Boston, MA 02115, USA
| | - Edward T Chouchani
- Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard University Medical School, Boston, MA 02115, USA
| | - Mark P Jedrychowski
- Department of Cell Biology, Harvard University Medical School, Boston, MA 02115, USA
| | - Brian K Erickson
- Department of Cell Biology, Harvard University Medical School, Boston, MA 02115, USA
| | - Kosaku Shinoda
- Diabetes Center, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA
| | - Paul Cohen
- Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard University Medical School, Boston, MA 02115, USA
| | - Ramalingam Vetrivelan
- Department of Neurology, Harvard Medical School and Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Gina Z Lu
- Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | - Sebastian C Hasenfuss
- Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard University Medical School, Boston, MA 02115, USA
| | - Shingo Kajimura
- Diabetes Center, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA
| | - Steve P Gygi
- Department of Cell Biology, Harvard University Medical School, Boston, MA 02115, USA
| | - Bruce M Spiegelman
- Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard University Medical School, Boston, MA 02115, USA.
| |
Collapse
|
428
|
Can thermogenic adipocytes protect from obesity? J Physiol Biochem 2015; 71:847-53. [PMID: 26482272 DOI: 10.1007/s13105-015-0443-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 10/08/2015] [Indexed: 10/22/2022]
Abstract
The role of brown adipocytes and adipocytes of a new beige type in the energy metabolism of a healthy person and in the pathogenesis of obesity has extensively been discussed in recent years. The interest to these cells has been stimulated owing to the application of new noninvasive methods for studying the metabolic activity of tissues. Using these methods, the presence of thermogenically active adipocytes in adults and their reactivity to cold stimuli have been proved. These data, together with the results of animal experiments support the idea of thermogenic fat being a direct regulator of the energy balance of man. However, for several reasons there are some objections to this viewpoint. The main objection is that the total activity of the human thermogenic adipocytes is about 100 kJ/day, i.e., it is negligible. In addition, the burn of excessive nutrients is biologically inappropriate for an organism. Therefore, the idea that obesity is caused by the decreased activity of thermogenic adipocytes is erroneous. The statement that the causes of obesity are associated with the increased efficiency of energy-dependent processes seems more reasonable. The consequence is a reduction in energy expenditure to perform a unit of biological work. This results in excess of nutrients deposited in the form of fat.
Collapse
|
429
|
Abstract
Since brown adipose tissue (BAT) dissipates energy through UCP1, BAT has garnered attention as a therapeutic intervention for obesity and metabolic diseases including type 2 diabetes. As we better understand the physiological roles of classical brown and beige adipocytes, it is becoming clear that BAT is not simply a heat-generating organ. Increased beige fat mass in response to a variety of external/internal cues is associated with significant improvements in glucose and lipid homeostasis that may not be entirely mediated by UCP1. We aim to discuss recent insights regarding the developmental lineages, molecular regulation, and new functions for brown and beige adipocytes.
Collapse
Affiliation(s)
- Shingo Kajimura
- UCSF Diabetes Center and Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143-0669, USA.
| | - Bruce M Spiegelman
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA.
| | - Patrick Seale
- Institute for Diabetes, Obesity, and Metabolism, Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
430
|
Seiler SE, Xu D, Ho JP, Lo KA, Buehrer BM, Ludlow YJW, Kovalik JP, Sun L. Characterization of a primary brown adipocyte culture system derived from human fetal interscapular fat. Adipocyte 2015; 4:303-10. [PMID: 26451287 DOI: 10.1080/21623945.2015.1042192] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 04/08/2015] [Accepted: 04/09/2015] [Indexed: 12/24/2022] Open
Abstract
Brown fat has gained widespread attention as a potential therapeutic target to treat obesity and associated metabolic disorders. Indeed, the anti-obesity potential of multiple targets to stimulate both brown adipocyte differentiation and recruitment have been verified in rodent models. However, their therapeutic potential in humans is unknown due to the lack of a human primary brown adipocyte cell culture system. Likewise, the lack of a well-characterized human model has limited the discovery of novel targets for the activation of human brown fat. To address this current need, we aimed to identify and describe the first primary brown adipocyte cell culture system from human fetal interscapular brown adipose tissue. Pre-adipocytes isolated from non-viable human fetal interscapular tissue were expanded and cryopreserved. Cells were then thawed and plated alongside adult human subcutaneous and omental pre-adipocytes for subsequent differentiation and phenotypic characterization. Interscapular pre-adipocytes in cell culture differentiated into mature adipocytes that were morphologically indistinguishable from the adult white depots. Throughout differentiation, cultured human fetal interscapular adipocytes demonstrated increased expression of classical brown fat markers compared to subcutaneous and omental cells. Further, functional analysis revealed an elevation in fatty acid oxidation as well as maximal and uncoupled oxygen consumption in interscapular brown adipocytes compared to white control cells. These data collectively identify the brown phenotype of these cells. Thus, our primary cell culture system derived from non-viable human fetal interscapular brown adipose tissue provides a valuable tool for the study of human brown adipocyte biology and for the development of anti-obesity therapeutics.
Collapse
|
431
|
Markan KR, Potthoff MJ. Metabolic fibroblast growth factors (FGFs): Mediators of energy homeostasis. Semin Cell Dev Biol 2015; 53:85-93. [PMID: 26428296 DOI: 10.1016/j.semcdb.2015.09.021] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 09/25/2015] [Indexed: 01/07/2023]
Abstract
The metabolic fibroblast growth factors (FGFs), FGF1, FGF15/19, and FGF21 differ from classic FGFs in that they modulate energy homeostasis in response to fluctuating nutrient availability. These unique mediators of metabolism regulate a number of physiological processes which contribute to their potent pharmacological properties. Administration of pharmacological doses of these FGFs causes weight loss, increases energy expenditure, and improves carbohydrate and lipid metabolism in obese animal models. However, many questions remain regarding the precise molecular and physiological mechanisms governing the effects of individual metabolic FGFs. Here we review the metabolic actions of FGF1, FGF15/19, and FGF21 while providing insights into their pharmacological effects by examining known biological functions.
Collapse
Affiliation(s)
- Kathleen R Markan
- Department of Pharmacology and University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Matthew J Potthoff
- Department of Pharmacology and University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| |
Collapse
|
432
|
Gorgey AS, Wells KM, Austin TL. Adiposity and spinal cord injury. World J Orthop 2015; 6:567-576. [PMID: 26396933 PMCID: PMC4573501 DOI: 10.5312/wjo.v6.i8.567] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 03/18/2015] [Accepted: 07/17/2015] [Indexed: 02/06/2023] Open
Abstract
The drastic changes in body composition following spinal cord injury (SCI) have been shown to play a significant role in cardiovascular and metabolic health. The pattern of storage and distribution of different types of adipose tissue may impact metabolic health variables similar to carbohydrate, lipid and bone metabolism. The use of magnetic resonance imaging provides insights on the interplay among different regional adipose tissue compartments and their role in developing chronic diseases. Regional adipose tissue can be either distributed centrally or peripherally into subcutaneous and ectopic sites. The primary ectopic adipose tissue sites are visceral, intramuscular and bone marrow. Dysfunction in the central nervous system following SCI impacts the pattern of distribution of adiposity especially between tetraplegia and paraplegia. The current editorial is focused primarily on introducing different types of adipose tissue and establishing scientific basis to develop appropriate dietary, rehabilitation or pharmaceutical interventions to manage the negative consequences of increasing adiposity after SCI. We have also summarized the clinical implications and future recommendations relevant to study adiposity after SCI.
Collapse
|
433
|
Claussnitzer M, Dankel SN, Kim KH, Quon G, Meuleman W, Haugen C, Glunk V, Sousa IS, Beaudry JL, Puviindran V, Abdennur NA, Liu J, Svensson PA, Hsu YH, Drucker DJ, Mellgren G, Hui CC, Hauner H, Kellis M. FTO Obesity Variant Circuitry and Adipocyte Browning in Humans. N Engl J Med 2015; 373:895-907. [PMID: 26287746 PMCID: PMC4959911 DOI: 10.1056/nejmoa1502214] [Citation(s) in RCA: 962] [Impact Index Per Article: 96.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Genomewide association studies can be used to identify disease-relevant genomic regions, but interpretation of the data is challenging. The FTO region harbors the strongest genetic association with obesity, yet the mechanistic basis of this association remains elusive. METHODS We examined epigenomic data, allelic activity, motif conservation, regulator expression, and gene coexpression patterns, with the aim of dissecting the regulatory circuitry and mechanistic basis of the association between the FTO region and obesity. We validated our predictions with the use of directed perturbations in samples from patients and from mice and with endogenous CRISPR-Cas9 genome editing in samples from patients. RESULTS Our data indicate that the FTO allele associated with obesity represses mitochondrial thermogenesis in adipocyte precursor cells in a tissue-autonomous manner. The rs1421085 T-to-C single-nucleotide variant disrupts a conserved motif for the ARID5B repressor, which leads to derepression of a potent preadipocyte enhancer and a doubling of IRX3 and IRX5 expression during early adipocyte differentiation. This results in a cell-autonomous developmental shift from energy-dissipating beige (brite) adipocytes to energy-storing white adipocytes, with a reduction in mitochondrial thermogenesis by a factor of 5, as well as an increase in lipid storage. Inhibition of Irx3 in adipose tissue in mice reduced body weight and increased energy dissipation without a change in physical activity or appetite. Knockdown of IRX3 or IRX5 in primary adipocytes from participants with the risk allele restored thermogenesis, increasing it by a factor of 7, and overexpression of these genes had the opposite effect in adipocytes from nonrisk-allele carriers. Repair of the ARID5B motif by CRISPR-Cas9 editing of rs1421085 in primary adipocytes from a patient with the risk allele restored IRX3 and IRX5 repression, activated browning expression programs, and restored thermogenesis, increasing it by a factor of 7. CONCLUSIONS Our results point to a pathway for adipocyte thermogenesis regulation involving ARID5B, rs1421085, IRX3, and IRX5, which, when manipulated, had pronounced pro-obesity and anti-obesity effects. (Funded by the German Research Center for Environmental Health and others.).
Collapse
Affiliation(s)
- Melina Claussnitzer
- From Beth Israel Deaconess Medical Center and Hebrew SeniorLife, Gerontology Division, Harvard Medical School, Boston (M.C., Y.-H.H.); Massachusetts Institute of Technology (MIT) Computer Science and Artificial Intelligence Laboratory (M.C., G.Q., W.M., N.A.A., M.K.), and Broad Institute of MIT and Harvard, Cambridge (M.C., G.Q., W.M., M.K.); Clinical Cooperation Group "Nutrigenomics and Type 2 Diabetes," Helmholtz Center Munich (M.C., H.H.), and Else Kröner-Fresenius Center for Nutritional Medicine, Klinikum rechts der Isar, ZIEL-Institute for Food and Health, Technische Universität München (M.C., V.G., I.S.S., H.H.), Munich, Germany; KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, and Hormone Laboratory, Haukeland University Hospital, Bergen, Norway (S.N.D., C.H., G.M.); Program in Developmental and Stem Cell Biology, Hospital for Sick Children, and Department of Molecular Genetics, University of Toronto (K.-H.K., V.P., J.L., C.-C.H.), and Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital (J.L.B., D.J.D.), Toronto; and the Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden (P.-A.S.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
434
|
Kato H, Tanaka G, Masuda S, Ogasawara J, Sakurai T, Kizaki T, Ohno H, Izawa T. Melatonin promotes adipogenesis and mitochondrial biogenesis in 3T3-L1 preadipocytes. J Pineal Res 2015; 59:267-75. [PMID: 26123001 DOI: 10.1111/jpi.12259] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 06/26/2015] [Indexed: 01/16/2023]
Abstract
Melatonin is synthesized in the pineal gland, but elicits a wide range of physiological responses in peripheral target tissues. Recent advances suggest that melatonin controls adiposity, resulting in changes in body weight. The aim of this study was to investigate the effect of melatonin on adipogenesis and mitochondrial biogenesis in 3T3-L1 mouse embryo fibroblasts. Melatonin significantly increased the expression of peroxisome proliferator-activated receptor-γ (PPAR-γ), a master regulator of adipogenesis, and promoted differentiation into adipocytes. Melatonin-treated cells also formed smaller lipid droplets and abundantly expressed several molecules associated with lipolysis, including adipose triglyceride lipase, perilipin, and comparative gene identification-58. Moreover, the hormone promoted biogenesis of mitochondria, as indicated by fluorescent staining, elevated the citrate synthase activity, and upregulated the expression of PPAR-γ coactivator 1 α, nuclear respiratory factor-1, and transcription factor A. The expression of uncoupling protein 1 was also observable both at mRNA and at protein level in melatonin-treated cells. Finally, adiponectin secretion and the expression of adiponectin receptors were enhanced. These results suggest that melatonin promotes adipogenesis, lipolysis, mitochondrial biogenesis, and adiponectin secretion. Thus, melatonin has potential as an anti-obesity agent that may reverse obesity-related disorders.
Collapse
Affiliation(s)
- Hisashi Kato
- Graduate School of Health and Sports Science, Doshisha University, Kyotanabe, Kyoto, Japan
| | - Goki Tanaka
- Graduate School of Health and Sports Science, Doshisha University, Kyotanabe, Kyoto, Japan
| | - Shinya Masuda
- Division of Diabetic Research, Clinical Research Institute, National Hospital Organization, Kyoto Medical Center, Fushimi, Kyoto, Japan
| | - Junetsu Ogasawara
- Department of Molecular Predictive Medicine and Sports Science, Kyorin University, Mitaka, Tokyo, Japan
| | - Takuya Sakurai
- Department of Molecular Predictive Medicine and Sports Science, Kyorin University, Mitaka, Tokyo, Japan
| | - Takako Kizaki
- Department of Molecular Predictive Medicine and Sports Science, Kyorin University, Mitaka, Tokyo, Japan
| | - Hideki Ohno
- Department of Molecular Predictive Medicine and Sports Science, Kyorin University, Mitaka, Tokyo, Japan
| | - Tetsuya Izawa
- Graduate School of Health and Sports Science, Doshisha University, Kyotanabe, Kyoto, Japan
- Faculty of Health and Sports Science, Doshisha University, Kyotanabe, Kyoto, Japan
| |
Collapse
|
435
|
Merlin J, Evans BA, Dehvari N, Sato M, Bengtsson T, Hutchinson DS. Could burning fat start with a brite spark? Pharmacological and nutritional ways to promote thermogenesis. Mol Nutr Food Res 2015. [DOI: 10.1002/mnfr.201500251] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jon Merlin
- Drug Discovery Biology; Monash Institute of Pharmaceutical Sciences; Monash University; Parkville Australia
| | - Bronwyn A. Evans
- Drug Discovery Biology; Monash Institute of Pharmaceutical Sciences; Monash University; Parkville Australia
| | - Nodi Dehvari
- Department of Molecular Biosciences; The Wenner-Gren Institute; Stockholm University; Stockholm Sweden
| | - Masaaki Sato
- Drug Discovery Biology; Monash Institute of Pharmaceutical Sciences; Monash University; Parkville Australia
- Department of Pharmacology; Monash University; Clayton Australia
| | - Tore Bengtsson
- Department of Molecular Biosciences; The Wenner-Gren Institute; Stockholm University; Stockholm Sweden
| | - Dana S. Hutchinson
- Drug Discovery Biology; Monash Institute of Pharmaceutical Sciences; Monash University; Parkville Australia
- Department of Pharmacology; Monash University; Clayton Australia
| |
Collapse
|
436
|
Sidossis LS, Porter C, Saraf MK, Børsheim E, Radhakrishnan RS, Chao T, Ali A, Chondronikola M, Mlcak R, Finnerty CC, Hawkins HK, Toliver-Kinsky T, Herndon DN. Browning of Subcutaneous White Adipose Tissue in Humans after Severe Adrenergic Stress. Cell Metab 2015; 22:219-27. [PMID: 26244931 PMCID: PMC4541608 DOI: 10.1016/j.cmet.2015.06.022] [Citation(s) in RCA: 312] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 10/24/2014] [Accepted: 06/23/2015] [Indexed: 01/04/2023]
Abstract
Since the presence of brown adipose tissue (BAT) was confirmed in adult humans, BAT has become a therapeutic target for obesity and insulin resistance. We examined whether human subcutaneous white adipose tissue (sWAT) can adopt a BAT-like phenotype using a clinical model of prolonged and severe adrenergic stress. sWAT samples were collected from severely burned and healthy individuals. A subset of burn victims were prospectively followed during their acute hospitalization. Browning of sWAT was determined by the presence of multilocular adipocytes, uncoupling protein 1 (UCP1), and increased mitochondrial density and respiratory capacity. Multilocular UCP1-positive adipocytes were found in sWAT samples from burn patients. UCP1 mRNA, mitochondrial density, and leak respiratory capacity in sWAT increased after burn trauma. Our data demonstrate that human sWAT can transform from an energy-storing to an energy-dissipating tissue, which opens new research avenues in our quest to prevent and treat obesity and its metabolic complications.
Collapse
Affiliation(s)
- Labros S Sidossis
- Metabolism Unit, Shriners Hospitals for Children - Galveston, 815 Market Street, Galveston, TX 77550, USA; Department of Nutrition and Dietetics, Harokopio University, El. Venizelou 70, Kallithea 176 76, Greece; Department of Internal Medicine, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA; Institute of Translational Sciences, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA; Department of Surgery, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA.
| | - Craig Porter
- Metabolism Unit, Shriners Hospitals for Children - Galveston, 815 Market Street, Galveston, TX 77550, USA; Department of Surgery, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA
| | - Manish K Saraf
- Metabolism Unit, Shriners Hospitals for Children - Galveston, 815 Market Street, Galveston, TX 77550, USA; Department of Surgery, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA
| | - Elisabet Børsheim
- Metabolism Unit, Shriners Hospitals for Children - Galveston, 815 Market Street, Galveston, TX 77550, USA; Department of Surgery, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA
| | - Ravi S Radhakrishnan
- Department of Surgery, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA
| | - Tony Chao
- Metabolism Unit, Shriners Hospitals for Children - Galveston, 815 Market Street, Galveston, TX 77550, USA; Department of Preventive Medicine and Community Healthy, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA
| | - Arham Ali
- Department of Surgery, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA
| | - Maria Chondronikola
- Metabolism Unit, Shriners Hospitals for Children - Galveston, 815 Market Street, Galveston, TX 77550, USA; Department of Preventive Medicine and Community Healthy, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA
| | - Ronald Mlcak
- Department of Surgery, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA
| | - Celeste C Finnerty
- Institute of Translational Sciences, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA; Department of Surgery, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA; Sealy Center for Molecular Medicine, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA
| | - Hal K Hawkins
- Department of Pathology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA
| | - Tracy Toliver-Kinsky
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA
| | - David N Herndon
- Metabolism Unit, Shriners Hospitals for Children - Galveston, 815 Market Street, Galveston, TX 77550, USA; Department of Surgery, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA
| |
Collapse
|
437
|
Of mice and men: novel insights regarding constitutive and recruitable brown adipocytes. INTERNATIONAL JOURNAL OF OBESITY SUPPLEMENTS 2015; 5:S15-20. [PMID: 27152169 DOI: 10.1038/ijosup.2015.5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 02/12/2015] [Indexed: 01/02/2023]
Abstract
Recently, there has been great attention given to the possibility of combating obesity by targeting brown fat activity or increasing differentiation of brown adipocytes in white fat depots through a process termed 'browning'. Sympathetic innervation of brown and white adipose tissues provides adrenergic input that drives thermogenesis and regulates fatty acid metabolism, as well as stimulating adipogenesis of recruitable brown adipocyte tissue (rBAT, also known as beige or brite) in white fat. Other factors acting in an endocrine or autocrine/paracrine manner in adipose tissue may also stimulate browning. There have been significant recent advances in understanding the mechanisms of increasing adipose tissue energy expenditure, as well as how brown adipocytes appear in white fat depots, including via de novo adipogenesis from tissue precursor cells. In this article, we integrate this new knowledge with a historical perspective on the discovery of 'browning'. We also provide an overview of constitutive BAT vs rBAT in mouse and human.
Collapse
|
438
|
Patil YN, Dille KN, Burk DH, Cortez CC, Gettys TW. Cellular and molecular remodeling of inguinal adipose tissue mitochondria by dietary methionine restriction. J Nutr Biochem 2015; 26:1235-47. [PMID: 26278039 DOI: 10.1016/j.jnutbio.2015.05.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 05/27/2015] [Accepted: 05/28/2015] [Indexed: 10/23/2022]
Abstract
Dietary methionine restriction (MR) produces a coordinated series of biochemical and physiological responses that improve biomarkers of metabolic health, increase energy expenditure, limit fat accretion and improve overall insulin sensitivity. Inguinal white adipose tissue (IWAT) is a primary target and site of action where the diet initiates transcriptional programs linked to enhancing both synthesis and oxidation of lipid. Using a combination of ex vivo approaches to assess dietary effects on cell morphology and function, we report that dietary MR produced a fourfold increase in multilocular, UCP1-expressing cells within this depot in conjunction with significant increases in mitochondrial content, size and cristae density. Dietary MR increased expression of multiple enzymes within the citric acid cycle, as well as respiratory complexes I, II and III. The physiological significance of these responses, evaluated in isolated mitochondria by high-resolution respirometry, was a significant increase in respiratory capacity measured using multiple substrates. The morphological, transcriptional and biochemical remodeling of IWAT mitochondria enhances the synthetic and oxidative capacity of this tissue and collectively underlies its expanded role as a significant contributor to the overall increase in metabolic flexibility and uncoupled respiration produced by the diet.
Collapse
Affiliation(s)
- Yuvraj N Patil
- Nutrient Sensing and Adipocyte Signaling Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | - Kelly N Dille
- Nutrient Sensing and Adipocyte Signaling Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | - David H Burk
- Nutrient Sensing and Adipocyte Signaling Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | - Cory C Cortez
- Nutrient Sensing and Adipocyte Signaling Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | - Thomas W Gettys
- Nutrient Sensing and Adipocyte Signaling Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808.
| |
Collapse
|
439
|
Hartig SM, Bader DA, Abadie KV, Motamed M, Hamilton MP, Long W, York B, Mueller M, Wagner M, Trauner M, Chan L, Bajaj M, Moore DD, Mancini MA, McGuire SE. Ubc9 Impairs Activation of the Brown Fat Energy Metabolism Program in Human White Adipocytes. Mol Endocrinol 2015; 29:1320-33. [PMID: 26192107 DOI: 10.1210/me.2015-1084] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Insulin resistance and type 2 diabetes mellitus (T2DM) result from an inability to efficiently store and catabolize surplus energy in adipose tissue. Subcutaneous adipocytes protect against insulin resistance and T2DM by coupling differentiation with the induction of brown fat gene programs for efficient energy metabolism. Mechanisms that disrupt these programs in adipocytes are currently poorly defined, but represent therapeutic targets for the treatment of T2DM. To gain insight into these mechanisms, we performed a high-throughput microscopy screen that identified ubiquitin carrier protein 9 (Ubc9) as a negative regulator of energy storage in human sc adipocytes. Ubc9 depletion enhanced energy storage and induced the brown fat gene program in human sc adipocytes. Induction of adipocyte differentiation resulted in decreased Ubc9 expression commensurate with increased brown fat gene expression. Thiazolidinedione treatment reduced the interaction between Ubc9 and peroxisome proliferator-activated receptor (PPAR)γ, suggesting a mechanism by which Ubc9 represses PPARγ activity. In support of this hypothesis, Ubc9 overexpression remodeled energy metabolism in human sc adipocytes by selectively inhibiting brown adipocyte-specific function. Further, Ubc9 overexpression decreased uncoupling protein 1 expression by disrupting PPARγ binding at a critical uncoupling protein 1 enhancer region. Last, Ubc9 is significantly elevated in sc adipose tissue isolated from mouse models of insulin resistance as well as diabetic and insulin-resistant humans. Taken together, our findings demonstrate a critical role for Ubc9 in the regulation of sc adipocyte energy homeostasis.
Collapse
Affiliation(s)
- Sean M Hartig
- Department of Molecular and Cellular Biology (S.M.H., D.A.B., K.V.A., M.Mo., M.P.H., W.L., B.Y., L.C., D.D.M., M.A.M., S.E.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Molecular Biology (W.L.), Wright State University Boonshoft School of Medicine, Dayton, Ohio 45435; Hans Popper Laboratory of Molecular Hepatology (M.Mu., M.T.), Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Laboratory of Experimental Hepatology (M.W.), Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Diabetes and Endocrinology Research Center (L.C., M.B.), Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, and the Baylor St Luke's Medical Center, Houston, Texas 77030; and Division of Radiation Oncology (S.E.M.), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - David A Bader
- Department of Molecular and Cellular Biology (S.M.H., D.A.B., K.V.A., M.Mo., M.P.H., W.L., B.Y., L.C., D.D.M., M.A.M., S.E.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Molecular Biology (W.L.), Wright State University Boonshoft School of Medicine, Dayton, Ohio 45435; Hans Popper Laboratory of Molecular Hepatology (M.Mu., M.T.), Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Laboratory of Experimental Hepatology (M.W.), Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Diabetes and Endocrinology Research Center (L.C., M.B.), Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, and the Baylor St Luke's Medical Center, Houston, Texas 77030; and Division of Radiation Oncology (S.E.M.), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Kathleen V Abadie
- Department of Molecular and Cellular Biology (S.M.H., D.A.B., K.V.A., M.Mo., M.P.H., W.L., B.Y., L.C., D.D.M., M.A.M., S.E.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Molecular Biology (W.L.), Wright State University Boonshoft School of Medicine, Dayton, Ohio 45435; Hans Popper Laboratory of Molecular Hepatology (M.Mu., M.T.), Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Laboratory of Experimental Hepatology (M.W.), Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Diabetes and Endocrinology Research Center (L.C., M.B.), Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, and the Baylor St Luke's Medical Center, Houston, Texas 77030; and Division of Radiation Oncology (S.E.M.), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Massoud Motamed
- Department of Molecular and Cellular Biology (S.M.H., D.A.B., K.V.A., M.Mo., M.P.H., W.L., B.Y., L.C., D.D.M., M.A.M., S.E.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Molecular Biology (W.L.), Wright State University Boonshoft School of Medicine, Dayton, Ohio 45435; Hans Popper Laboratory of Molecular Hepatology (M.Mu., M.T.), Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Laboratory of Experimental Hepatology (M.W.), Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Diabetes and Endocrinology Research Center (L.C., M.B.), Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, and the Baylor St Luke's Medical Center, Houston, Texas 77030; and Division of Radiation Oncology (S.E.M.), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Mark P Hamilton
- Department of Molecular and Cellular Biology (S.M.H., D.A.B., K.V.A., M.Mo., M.P.H., W.L., B.Y., L.C., D.D.M., M.A.M., S.E.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Molecular Biology (W.L.), Wright State University Boonshoft School of Medicine, Dayton, Ohio 45435; Hans Popper Laboratory of Molecular Hepatology (M.Mu., M.T.), Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Laboratory of Experimental Hepatology (M.W.), Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Diabetes and Endocrinology Research Center (L.C., M.B.), Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, and the Baylor St Luke's Medical Center, Houston, Texas 77030; and Division of Radiation Oncology (S.E.M.), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Weiwen Long
- Department of Molecular and Cellular Biology (S.M.H., D.A.B., K.V.A., M.Mo., M.P.H., W.L., B.Y., L.C., D.D.M., M.A.M., S.E.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Molecular Biology (W.L.), Wright State University Boonshoft School of Medicine, Dayton, Ohio 45435; Hans Popper Laboratory of Molecular Hepatology (M.Mu., M.T.), Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Laboratory of Experimental Hepatology (M.W.), Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Diabetes and Endocrinology Research Center (L.C., M.B.), Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, and the Baylor St Luke's Medical Center, Houston, Texas 77030; and Division of Radiation Oncology (S.E.M.), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Brian York
- Department of Molecular and Cellular Biology (S.M.H., D.A.B., K.V.A., M.Mo., M.P.H., W.L., B.Y., L.C., D.D.M., M.A.M., S.E.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Molecular Biology (W.L.), Wright State University Boonshoft School of Medicine, Dayton, Ohio 45435; Hans Popper Laboratory of Molecular Hepatology (M.Mu., M.T.), Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Laboratory of Experimental Hepatology (M.W.), Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Diabetes and Endocrinology Research Center (L.C., M.B.), Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, and the Baylor St Luke's Medical Center, Houston, Texas 77030; and Division of Radiation Oncology (S.E.M.), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Michaela Mueller
- Department of Molecular and Cellular Biology (S.M.H., D.A.B., K.V.A., M.Mo., M.P.H., W.L., B.Y., L.C., D.D.M., M.A.M., S.E.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Molecular Biology (W.L.), Wright State University Boonshoft School of Medicine, Dayton, Ohio 45435; Hans Popper Laboratory of Molecular Hepatology (M.Mu., M.T.), Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Laboratory of Experimental Hepatology (M.W.), Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Diabetes and Endocrinology Research Center (L.C., M.B.), Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, and the Baylor St Luke's Medical Center, Houston, Texas 77030; and Division of Radiation Oncology (S.E.M.), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Martin Wagner
- Department of Molecular and Cellular Biology (S.M.H., D.A.B., K.V.A., M.Mo., M.P.H., W.L., B.Y., L.C., D.D.M., M.A.M., S.E.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Molecular Biology (W.L.), Wright State University Boonshoft School of Medicine, Dayton, Ohio 45435; Hans Popper Laboratory of Molecular Hepatology (M.Mu., M.T.), Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Laboratory of Experimental Hepatology (M.W.), Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Diabetes and Endocrinology Research Center (L.C., M.B.), Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, and the Baylor St Luke's Medical Center, Houston, Texas 77030; and Division of Radiation Oncology (S.E.M.), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Michael Trauner
- Department of Molecular and Cellular Biology (S.M.H., D.A.B., K.V.A., M.Mo., M.P.H., W.L., B.Y., L.C., D.D.M., M.A.M., S.E.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Molecular Biology (W.L.), Wright State University Boonshoft School of Medicine, Dayton, Ohio 45435; Hans Popper Laboratory of Molecular Hepatology (M.Mu., M.T.), Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Laboratory of Experimental Hepatology (M.W.), Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Diabetes and Endocrinology Research Center (L.C., M.B.), Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, and the Baylor St Luke's Medical Center, Houston, Texas 77030; and Division of Radiation Oncology (S.E.M.), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Lawrence Chan
- Department of Molecular and Cellular Biology (S.M.H., D.A.B., K.V.A., M.Mo., M.P.H., W.L., B.Y., L.C., D.D.M., M.A.M., S.E.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Molecular Biology (W.L.), Wright State University Boonshoft School of Medicine, Dayton, Ohio 45435; Hans Popper Laboratory of Molecular Hepatology (M.Mu., M.T.), Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Laboratory of Experimental Hepatology (M.W.), Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Diabetes and Endocrinology Research Center (L.C., M.B.), Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, and the Baylor St Luke's Medical Center, Houston, Texas 77030; and Division of Radiation Oncology (S.E.M.), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Mandeep Bajaj
- Department of Molecular and Cellular Biology (S.M.H., D.A.B., K.V.A., M.Mo., M.P.H., W.L., B.Y., L.C., D.D.M., M.A.M., S.E.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Molecular Biology (W.L.), Wright State University Boonshoft School of Medicine, Dayton, Ohio 45435; Hans Popper Laboratory of Molecular Hepatology (M.Mu., M.T.), Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Laboratory of Experimental Hepatology (M.W.), Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Diabetes and Endocrinology Research Center (L.C., M.B.), Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, and the Baylor St Luke's Medical Center, Houston, Texas 77030; and Division of Radiation Oncology (S.E.M.), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - David D Moore
- Department of Molecular and Cellular Biology (S.M.H., D.A.B., K.V.A., M.Mo., M.P.H., W.L., B.Y., L.C., D.D.M., M.A.M., S.E.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Molecular Biology (W.L.), Wright State University Boonshoft School of Medicine, Dayton, Ohio 45435; Hans Popper Laboratory of Molecular Hepatology (M.Mu., M.T.), Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Laboratory of Experimental Hepatology (M.W.), Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Diabetes and Endocrinology Research Center (L.C., M.B.), Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, and the Baylor St Luke's Medical Center, Houston, Texas 77030; and Division of Radiation Oncology (S.E.M.), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Michael A Mancini
- Department of Molecular and Cellular Biology (S.M.H., D.A.B., K.V.A., M.Mo., M.P.H., W.L., B.Y., L.C., D.D.M., M.A.M., S.E.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Molecular Biology (W.L.), Wright State University Boonshoft School of Medicine, Dayton, Ohio 45435; Hans Popper Laboratory of Molecular Hepatology (M.Mu., M.T.), Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Laboratory of Experimental Hepatology (M.W.), Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Diabetes and Endocrinology Research Center (L.C., M.B.), Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, and the Baylor St Luke's Medical Center, Houston, Texas 77030; and Division of Radiation Oncology (S.E.M.), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Sean E McGuire
- Department of Molecular and Cellular Biology (S.M.H., D.A.B., K.V.A., M.Mo., M.P.H., W.L., B.Y., L.C., D.D.M., M.A.M., S.E.M.), Baylor College of Medicine, Houston, Texas 77030; Department of Biochemistry and Molecular Biology (W.L.), Wright State University Boonshoft School of Medicine, Dayton, Ohio 45435; Hans Popper Laboratory of Molecular Hepatology (M.Mu., M.T.), Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; Laboratory of Experimental Hepatology (M.W.), Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Diabetes and Endocrinology Research Center (L.C., M.B.), Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Baylor College of Medicine, and the Baylor St Luke's Medical Center, Houston, Texas 77030; and Division of Radiation Oncology (S.E.M.), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| |
Collapse
|
440
|
Jankovic A, Golic I, Markelic M, Stancic A, Otasevic V, Buzadzic B, Korac A, Korac B. Two key temporally distinguishable molecular and cellular components of white adipose tissue browning during cold acclimation. J Physiol 2015; 593:3267-80. [PMID: 26096127 DOI: 10.1113/jp270805] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/08/2015] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS White to brown adipose tissue conversion and thermogenesis can be ignited by different conditions or agents and its sustainability over the long term is still unclear. Browning of rat retroperitoneal white adipose tissue (rpWAT) during cold acclimation involves two temporally apparent components: (1) a predominant non-selective browning of most adipocytes and an initial sharp but transient induction of uncoupling protein 1, peroxisome proliferator-activated receptor (PPAR) coactivator-1α, PPARγ and PPARα expression, and (2) the subsistence of relatively few thermogenically competent adipocytes after 45 days of cold acclimation. The different behaviours of two rpWAT beige/brown adipocyte subsets control temporal aspects of the browning process, and thus regulation of both components may influence body weight and the potential successfulness of anti-obesity therapies. ABSTRACT Conversion of white into brown adipose tissue may have important implications in obesity resistance and treatment. Several browning agents or conditions ignite thermogenesis in white adipose tissue (WAT). To reveal the capacity of WAT to function in a brownish/burning mode over the long term, we investigated the progression of the rat retroperitoneal WAT (rpWAT) browning during 45 days of cold acclimation. During the early stages of cold acclimation, the majority of rpWAT adipocytes underwent multilocularization and thermogenic-profile induction, as demonstrated by the presence of a multitude of uncoupling protein 1 (UCP1)-immunopositive paucilocular adipocytes containing peroxisome proliferator-activated receptor (PPAR) coactivator-1α (PGC-1α) and PR domain-containing 16 (PRDM16) in their nuclei. After 45 days, all adipocytes remained PRDM16 immunopositive, but only a few multilocular adipocytes rich in mitochondria remained UCP1/PGC-1α immunopositive. Molecular evidence showed that thermogenic recruitment of rpWAT occurred following cold exposure, but returned to starting levels after cold acclimation. Compared with controls (22 ± 1 °C), levels of UCP1 mRNA increased in parallel with PPARγ (PPARα from days 1 to 7 and PGC-1α on day 1). Transcriptional recruitment of rpWAT was followed by an increase in UCP1 protein content (from days 1 to 21). Results clearly showed that most of the adipocytes within rpWAT underwent transient brown-fat-like thermogenic recruitment upon stimulation, but only a minority of cells retained a brown adipose tissue-like phenotype after the attainment of cold acclimation. Therefore, browning of WAT is dependent on both maintaining the thermogenic response and retaining enough brown-like thermogenically competent adipocytes in the long-term. Both aspects of browning could be important for long-term energy homeostasis and body-weight regulation.
Collapse
Affiliation(s)
- Aleksandra Jankovic
- Department of Physiology, Institute for Biological Research 'Sinisa Stankovic', University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Igor Golic
- Faculty of Biology, Center for Electron Microscopy, University of Belgrade, Studentski trg 16, 11000, Belgrade, Serbia
| | - Milica Markelic
- Faculty of Biology, Center for Electron Microscopy, University of Belgrade, Studentski trg 16, 11000, Belgrade, Serbia
| | - Ana Stancic
- Department of Physiology, Institute for Biological Research 'Sinisa Stankovic', University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Vesna Otasevic
- Department of Physiology, Institute for Biological Research 'Sinisa Stankovic', University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Biljana Buzadzic
- Department of Physiology, Institute for Biological Research 'Sinisa Stankovic', University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Aleksandra Korac
- Faculty of Biology, Center for Electron Microscopy, University of Belgrade, Studentski trg 16, 11000, Belgrade, Serbia
| | - Bato Korac
- Department of Physiology, Institute for Biological Research 'Sinisa Stankovic', University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| |
Collapse
|
441
|
Cohen P, Spiegelman BM. Brown and Beige Fat: Molecular Parts of a Thermogenic Machine. Diabetes 2015; 64:2346-51. [PMID: 26050670 PMCID: PMC4477363 DOI: 10.2337/db15-0318] [Citation(s) in RCA: 206] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 03/17/2015] [Indexed: 12/17/2022]
Abstract
The epidemic of obesity and type 2 diabetes has increased interest in pathways that affect energy balance in mammalian systems. Brown fat, in all of its dimensions, can increase energy expenditure through the dissipation of chemical energy in the form of heat, using mitochondrial uncoupling and perhaps other pathways. We discuss here some of the thermodynamic and cellular aspects of recent progress in brown fat research. This includes studies of developmental lineages of UCP1(+) adipocytes, including the discovery of beige fat cells, a new thermogenic cell type. We also discuss the physiology and transcriptional control of brown and beige cells in rodents and the state of current knowledge about human brown fat.
Collapse
Affiliation(s)
- Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY
| | - Bruce M Spiegelman
- Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, MA
| |
Collapse
|
442
|
Douris N, Stevanovic DM, Fisher FM, Cisu TI, Chee MJ, Nguyen NL, Zarebidaki E, Adams AC, Kharitonenkov A, Flier JS, Bartness TJ, Maratos-Flier E. Central Fibroblast Growth Factor 21 Browns White Fat via Sympathetic Action in Male Mice. Endocrinology 2015; 156:2470-81. [PMID: 25924103 PMCID: PMC4475718 DOI: 10.1210/en.2014-2001] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Fibroblast growth factor 21 (FGF21) has multiple metabolic actions, including the induction of browning in white adipose tissue. Although FGF21 stimulated browning results from a direct interaction between FGF21 and the adipocyte, browning is typically associated with activation of the sympathetic nervous system through cold exposure. We tested the hypothesis that FGF21 can act via the brain, to increase sympathetic activity and induce browning, independent of cell-autonomous actions. We administered FGF21 into the central nervous system via lateral ventricle infusion into male mice and found that the central treatment increased norepinephrine turnover in target tissues that include the inguinal white adipose tissue and brown adipose tissue. Central FGF21 stimulated browning as assessed by histology, expression of uncoupling protein 1, and the induction of gene expression associated with browning. These effects were markedly attenuated when mice were treated with a β-blocker. Additionally, neither centrally nor peripherally administered FGF21 initiated browning in mice lacking β-adrenoceptors, demonstrating that an intact adrenergic system is necessary for FGF21 action. These data indicate that FGF21 can signal in the brain to activate the sympathetic nervous system and induce adipose tissue thermogenesis.
Collapse
MESH Headings
- Adipocytes, Brown/metabolism
- Adipocytes, White/drug effects
- Adipocytes, White/metabolism
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/metabolism
- Adrenergic beta-Antagonists/pharmacology
- Animals
- Fibroblast Growth Factors/pharmacology
- Infusions, Intraventricular
- Ion Channels/drug effects
- Ion Channels/metabolism
- Male
- Mice
- Mice, Knockout
- Mitochondrial Proteins/drug effects
- Mitochondrial Proteins/metabolism
- Receptors, Adrenergic, beta/genetics
- Receptors, Adrenergic, beta-1/genetics
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-3/genetics
- Sympathetic Nervous System/drug effects
- Sympathetic Nervous System/metabolism
- Thermogenesis
- Uncoupling Protein 1
Collapse
Affiliation(s)
- Nicholas Douris
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Darko M Stevanovic
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Ffolliott M Fisher
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Theodore I Cisu
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Melissa J Chee
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Ngoc L Nguyen
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Eleen Zarebidaki
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Andrew C Adams
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Alexei Kharitonenkov
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Jeffrey S Flier
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Timothy J Bartness
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| | - Eleftheria Maratos-Flier
- Division of Endocrinology (N.D., D.M.S., f.M.F., T.I.C., M.J.C., J.S.F., E.M.-F.), Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215-5491; Institute of Medical Physiology (D.M.S.), School of Medicine, University of Belgrade, 11000 Belgrade, Serbia; Department of Biology and Center for Obesity Reversal (N.L.N., E.Z., T.J.B.), Georgia State University, Atlanta, Georgia 30302-4010; and Diabetes Research (A.C.A., A.K.), Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285-0001
| |
Collapse
|
443
|
Lizárraga-Mollinedo E, Fernández-Millán E, García-San Frutos M, de Toro-Martín J, Fernández-Agulló T, Ros M, Álvarez C, Escrivá F. Early and Long-term Undernutrition in Female Rats Exacerbates the Metabolic Risk Associated with Nutritional Rehabilitation. J Biol Chem 2015; 290:19353-66. [PMID: 26105051 DOI: 10.1074/jbc.m114.549204] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Indexed: 12/23/2022] Open
Abstract
Human studies have suggested that early undernutrition increases the risk of obesity, thereby explaining the increase in overweight among individuals from developing countries who have been undernourished as children. However, this conclusion is controversial, given that other studies do not concur. This study sought to determine whether rehabilitation after undernutrition increases the risk of obesity and metabolic disorders. We employed a published experimental food-restriction model. Wistar female rats subjected to severe food restriction since fetal stage and controls were transferred to a moderately high-fat diet (cafeteria) provided at 70 days of life to 6.5 months. Another group of undernourished rats were rehabilitated with chow. The energy intake of undernourished animals transferred to cafeteria formula exceeded that of the controls under this regime and was probably driven by hypothalamic disorders in insulin and leptin signal transduction. The cafeteria diet resulted in greater relative increases in both fat and lean body mass in the undernourished rats when compared with controls, enabling the former group to completely catch up in length and body mass index. White adipose tissues of undernourished rats transferred to the high-lipid regime developed a browning which, probably, contributed to avoid the obesigenic effect observed in controls. Nevertheless, the restricted group rehabilitated with cafeteria formula had greater accretion of visceral than subcutaneous fat, showed increased signs of macrophage infiltration and inflammation in visceral pad, dyslipidemia, and ectopic fat accumulation. The data indicate that early long-term undernutrition is associated with increased susceptibility to the harmful effects of nutritional rehabilitation, without causing obesity.
Collapse
Affiliation(s)
- Esther Lizárraga-Mollinedo
- From the Departamento de Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain, the Centro de Investigación en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, 28029 Spain, and
| | - Elisa Fernández-Millán
- the Centro de Investigación en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, 28029 Spain, and
| | - Miriam García-San Frutos
- the Departamento de Bioquímica, Fisiología y Genética Molecular, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, 28922 Madrid, Spain
| | - Juan de Toro-Martín
- From the Departamento de Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain
| | - Teresa Fernández-Agulló
- the Departamento de Bioquímica, Fisiología y Genética Molecular, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, 28922 Madrid, Spain
| | - Manuel Ros
- the Departamento de Bioquímica, Fisiología y Genética Molecular, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, 28922 Madrid, Spain
| | - Carmen Álvarez
- From the Departamento de Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain, the Centro de Investigación en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, 28029 Spain, and
| | - Fernando Escrivá
- From the Departamento de Bioquímica y Biología Molecular II, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain, the Centro de Investigación en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, 28029 Spain, and
| |
Collapse
|
444
|
de Jong JMA, Larsson O, Cannon B, Nedergaard J. A stringent validation of mouse adipose tissue identity markers. Am J Physiol Endocrinol Metab 2015; 308:E1085-105. [PMID: 25898951 DOI: 10.1152/ajpendo.00023.2015] [Citation(s) in RCA: 222] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 04/15/2015] [Indexed: 12/28/2022]
Abstract
The nature of brown adipose tissue in humans is presently debated: whether it is classical brown or of brite/beige nature. The dissimilar developmental origins and proposed distinct functions of the brown and brite/beige tissues make it essential to ascertain the identity of human depots with the perspective of recruiting and activating them for the treatment of obesity and type 2 diabetes. For identification of the tissues, a number of marker genes have been proposed, but the validity of the markers has not been well documented. We used established brown (interscapular), brite (inguinal), and white (epididymal) mouse adipose tissues and corresponding primary cell cultures as validators and examined the informative value of a series of suggested markers earlier used in the discussion considering the nature of human brown adipose tissue. Most of these markers unexpectedly turned out to be noninformative concerning tissue classification (Car4, Cited1, Ebf3, Eva1, Fbxo31, Fgf21, Lhx8, Hoxc8, and Hoxc9). Only Zic1 (brown), Cd137, Epsti1, Tbx1, Tmem26 (brite), and Tcf21 (white) proved to be informative in these three tissues. However, the expression of the brite markers was not maintained in cell culture. In a more extensive set of adipose depots, these validated markers provide new information about depot identity. Principal component analysis supported our single-gene conclusions. Furthermore, Zic1, Hoxc8, Hoxc9, and Tcf21 displayed anteroposterior expression patterns, indicating a relationship between anatomic localization and adipose tissue identity (and possibly function). Together, the observed expression patterns of these validated marker genes necessitates reconsideration of adipose depot identity in mice and humans.
Collapse
Affiliation(s)
- Jasper M A de Jong
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden; and
| | - Ola Larsson
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden; and
| | - Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden; and
| |
Collapse
|
445
|
Shabalina IG, Kalinovich AV, Cannon B, Nedergaard J. Metabolically inert perfluorinated fatty acids directly activate uncoupling protein 1 in brown-fat mitochondria. Arch Toxicol 2015; 90:1117-28. [PMID: 26041126 PMCID: PMC4830884 DOI: 10.1007/s00204-015-1535-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 05/12/2015] [Indexed: 12/14/2022]
Abstract
The metabolically inert perfluorinated fatty acids perfluorooctane sulfonate (PFOS) and perfluorooctanoate (PFOA) can display fatty acid-like activity in biological systems. The uncoupling protein 1 (UCP1) in brown adipose tissue is physiologically (re)activated by fatty acids, including octanoate. This leads to bioenergetically uncoupled energy dissipation (heat production, thermogenesis). We have examined here the possibility that PFOA/PFOS can directly (re)activate UCP1 in isolated mouse brown-fat mitochondria. In wild-type brown-fat mitochondria, PFOS and PFOA overcame GDP-inhibited thermogenesis, leading to increased oxygen consumption and dissipated membrane potential. The absence of this effect in brown-fat mitochondria from UCP1-ablated mice indicated that it occurred through activation of UCP1. A competitive type of inhibition by increased GDP concentrations indicated interaction with the same mechanistic site as that utilized by fatty acids. No effect was observed in heart mitochondria, i.e., in mitochondria without UCP1. The stimulatory effect of PFOA/PFOS was not secondary to non-specific mitochondrial membrane permeabilization or to ROS production. Thus, metabolic effects of perfluorinated fatty acids could include direct brown adipose tissue (UCP1) activation. The possibility that this may lead to unwarranted extra heat production and thus extra utilization of food resources, leading to decreased fitness in mammalian wildlife, is discussed, as well as possible negative effects in humans. However, a possibility to utilize PFOA-/PFOS-like substances for activating UCP1 therapeutically in obesity-prone humans may also be envisaged.
Collapse
Affiliation(s)
- Irina G Shabalina
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, SE-106 91, Stockholm, Sweden
| | - Anastasia V Kalinovich
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, SE-106 91, Stockholm, Sweden
| | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, SE-106 91, Stockholm, Sweden
| | - Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, SE-106 91, Stockholm, Sweden.
| |
Collapse
|
446
|
Stanford KI, Middelbeek RJW, Townsend KL, Lee MY, Takahashi H, So K, Hitchcox KM, Markan KR, Hellbach K, Hirshman MF, Tseng YH, Goodyear LJ. A novel role for subcutaneous adipose tissue in exercise-induced improvements in glucose homeostasis. Diabetes 2015; 64:2002-14. [PMID: 25605808 PMCID: PMC4439563 DOI: 10.2337/db14-0704] [Citation(s) in RCA: 233] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 01/10/2015] [Indexed: 01/10/2023]
Abstract
Exercise training improves whole-body glucose homeostasis through effects largely attributed to adaptations in skeletal muscle; however, training also affects other tissues, including adipose tissue. To determine whether exercise-induced adaptations to adipose tissue contribute to training-induced improvements in glucose homeostasis, subcutaneous white adipose tissue (scWAT) from exercise-trained or sedentary donor mice was transplanted into the visceral cavity of sedentary recipients. Remarkably, 9 days post-transplantation, mice receiving scWAT from exercise-trained mice had improved glucose tolerance and enhanced insulin sensitivity compared with mice transplanted with scWAT from sedentary or sham-treated mice. Mice transplanted with scWAT from exercise-trained mice had increased insulin-stimulated glucose uptake in tibialis anterior and soleus muscles and brown adipose tissue, suggesting that the transplanted scWAT exerted endocrine effects. Furthermore, the deleterious effects of high-fat feeding on glucose tolerance and insulin sensitivity were completely reversed if high-fat-fed recipient mice were transplanted with scWAT from exercise-trained mice. In additional experiments, voluntary exercise training by wheel running for only 11 days resulted in profound changes in scWAT, including the increased expression of ∼1,550 genes involved in numerous cellular functions including metabolism. Exercise training causes adaptations to scWAT that elicit metabolic improvements in other tissues, demonstrating a previously unrecognized role for adipose tissue in the beneficial effects of exercise on systemic glucose homeostasis.
Collapse
Affiliation(s)
- Kristin I Stanford
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Roeland J W Middelbeek
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Kristy L Townsend
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Min-Young Lee
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hirokazu Takahashi
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Kawai So
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Kristen M Hitchcox
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Kathleen R Markan
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Katharina Hellbach
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Michael F Hirshman
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
447
|
Chondronikola M, Porter C, Lidell ME, Sidossis LS. Response to comment on Chondronikola et al. Brown adipose tissue improves whole-body glucose homeostasis and insulin sensitivity in humans. Diabetes 2014;63:4089-4099. Diabetes 2015; 64:e14-5. [PMID: 25999542 DOI: 10.2337/db15-0147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Maria Chondronikola
- Metabolism Unit, Shriners Hospital for Children, Galveston, TX Department of Preventive Medicine and Community Health, University of Texas Medical Branch, Galveston, TX Department of Nutrition and Metabolism, Division of Rehabilitation Sciences, University of Texas Medical Branch, Galveston, TX Department of Nutrition and Dietetics, Harokopio University of Athens, Athens, Greece
| | - Craig Porter
- Metabolism Unit, Shriners Hospital for Children, Galveston, TX Department of Surgery, University of Texas Medical Branch, Galveston, TX
| | - Martin E Lidell
- Department of Medical and Clinical Genetics, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Labros S Sidossis
- Metabolism Unit, Shriners Hospital for Children, Galveston, TX Department of Nutrition and Metabolism, Division of Rehabilitation Sciences, University of Texas Medical Branch, Galveston, TX Department of Nutrition and Dietetics, Harokopio University of Athens, Athens, Greece Department of Surgery, University of Texas Medical Branch, Galveston, TX Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX Sealy Center on Aging, University of Texas Medical Branch, Galveston, TX Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX
| |
Collapse
|
448
|
Brestoff JR, Artis D. Immune regulation of metabolic homeostasis in health and disease. Cell 2015; 161:146-160. [PMID: 25815992 DOI: 10.1016/j.cell.2015.02.022] [Citation(s) in RCA: 352] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 01/27/2015] [Accepted: 01/28/2015] [Indexed: 02/07/2023]
Abstract
Obesity is an increasingly prevalent disease worldwide. While genetic and environmental factors are known to regulate the development of obesity and associated metabolic diseases, emerging studies indicate that innate and adaptive immune cell responses in adipose tissue have critical roles in the regulation of metabolic homeostasis. In the lean state, type 2 cytokine-associated immune cell responses predominate in white adipose tissue and protect against weight gain and insulin resistance through direct effects on adipocytes and elicitation of beige adipose. In obesity, these metabolically beneficial immune pathways become dysregulated, and adipocytes and other factors initiate metabolically deleterious type 1 inflammation that impairs glucose metabolism. This review discusses our current understanding of the functions of different types of adipose tissue and how immune cells regulate adipocyte function and metabolic homeostasis in the context of health and disease and highlights. We also highlight the potential of targeting immuno-metabolic pathways as a therapeutic strategy to treat obesity and associated diseases.
Collapse
Affiliation(s)
- Jonathan R Brestoff
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, NY, 10021, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, NY, 10021, USA.
| |
Collapse
|
449
|
Shabalina IG, Kramarova TV, Mattsson CL, Petrovic N, Rahman Qazi M, Csikasz RI, Chang SC, Butenhoff J, DePierre JW, Cannon B, Nedergaard J. The Environmental Pollutants Perfluorooctane Sulfonate and Perfluorooctanoic Acid Upregulate Uncoupling Protein 1 (UCP1) in Brown-Fat Mitochondria Through a UCP1-Dependent Reduction in Food Intake. Toxicol Sci 2015; 146:334-43. [PMID: 26001964 DOI: 10.1093/toxsci/kfv098] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The environmental pollutants perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA) cause a dramatic reduction in the size of the major adipose tissue depots and a general body weight decrease when they are added to the food of mice. We demonstrate here that this is mainly due to a reduction in food intake; this reduction was not due to food aversion. Remarkably and unexpectedly, a large part of the effect of PFOA/PFOS on food intake was dependent on the presence of the uncoupling protein 1 (UCP1) in the mice. Correspondingly, PFOA/PFOS treatment induced recruitment of brown adipose tissue mitochondria: increased oxidative capacity and increased UCP1-mediated oxygen consumption (thermogenesis). In mice pair-fed to the food intake during PFOA/PFOS treatment in wildtype mice, brown-fat mitochondrial recruitment was also induced. We conclude that we have uncovered the existence of a regulatory component of food intake that is dependent upon brown adipose tissue thermogenic activity. The possible environmental consequences of this novel PFOA/PFOS effect (a possible decreased fitness) are noted, as well as the perspectives of this finding on the general understanding of control of food intake control and its possible extension to combatting obesity.
Collapse
Affiliation(s)
| | | | | | - Natasa Petrovic
- *Department of Molecular Biosciences, The Wenner-Gren Institute
| | - Mousumi Rahman Qazi
- The Arrhenius Laboratories F3, Department of Biochemistry and Biophysics, Stockholm University, SE-106 91 Stockholm, Sweden; and
| | | | | | - John Butenhoff
- Medical Department, 3 M Center, St. Paul, Minnesota 55144
| | - Joseph W DePierre
- The Arrhenius Laboratories F3, Department of Biochemistry and Biophysics, Stockholm University, SE-106 91 Stockholm, Sweden; and
| | - Barbara Cannon
- *Department of Molecular Biosciences, The Wenner-Gren Institute
| | - Jan Nedergaard
- *Department of Molecular Biosciences, The Wenner-Gren Institute;
| |
Collapse
|
450
|
Keipert S, Kutschke M, Lamp D, Brachthäuser L, Neff F, Meyer CW, Oelkrug R, Kharitonenkov A, Jastroch M. Genetic disruption of uncoupling protein 1 in mice renders brown adipose tissue a significant source of FGF21 secretion. Mol Metab 2015; 4:537-42. [PMID: 26137441 PMCID: PMC4481421 DOI: 10.1016/j.molmet.2015.04.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 04/27/2015] [Accepted: 04/30/2015] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE Circulating fibroblast growth factor 21 (FGF21) is an important auto- and endocrine player with beneficial metabolic effects on obesity and diabetes. In humans, thermogenic brown adipose tissue (BAT) was recently suggested as a source of FGF21 secretion during cold exposure. Here, we aim to clarify the role of UCP1 and ambient temperature in the regulation of FGF21 in mice. METHODS Wildtype (WT) and UCP1-knockout (UCP1 KO) mice, the latter being devoid of BAT-derived non-shivering thermogenesis, were exposed to different housing temperatures. Plasma metabolites and FGF21 levels were determined, gene expression was analyzed by qPCR, and tissue histology was performed with adipose tissue. RESULTS At thermoneutrality, FGF21 gene expression and serum levels were not different between WT and UCP1 KO mice. Cold exposure led to highly increased FGF21 serum levels in UCP1 KO mice, which were reflected in increased FGF21 gene expression in adipose tissues but not in liver and skeletal muscle. Ex vivo secretion assays revealed FGF21 release only from BAT, progressively increasing with decreasing ambient temperatures. In association with increased FGF21 serum levels in the UCP1 KO mouse, typical FGF21-related serum metabolites and inguinal white adipose tissue morphology and thermogenic gene expression were altered. CONCLUSIONS Here we show that the genetic ablation of UCP1 increases FGF21 gene expression in adipose tissue. The removal of adaptive nonshivering thermogenesis renders BAT a significant source of endogenous FGF21 under thermal stress. Thus, the thermogenic competence of BAT is not a requirement for FGF21 secretion. Notably, high endogenous FGF21 levels in UCP1-deficient models and subjects may confound pharmacological FGF21 treatments.
Collapse
Affiliation(s)
- Susanne Keipert
- Helmholtz Diabetes Center, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Maria Kutschke
- Helmholtz Diabetes Center, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Daniel Lamp
- Helmholtz Diabetes Center, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Laura Brachthäuser
- Institute of Pathology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Frauke Neff
- Institute of Pathology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Carola W. Meyer
- Helmholtz Diabetes Center, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Animal Physiology, Philipps-Universität, 35043 Marburg, Germany
| | - Rebecca Oelkrug
- Institute of Pharmacology and Toxicology, Biomedical Center, University of Bonn, 53105 Bonn, Germany
- Department of Animal Physiology, Philipps-Universität, 35043 Marburg, Germany
| | | | - Martin Jastroch
- Helmholtz Diabetes Center, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Animal Physiology, Philipps-Universität, 35043 Marburg, Germany
- Corresponding author. Helmholtz Diabetes Center, Helmholtz Zentrum München, 85764 Neuherberg, Germany. Tel.: +49 89 3187 2105.
| |
Collapse
|