1
|
Barsky ST, Monks DA. Lifespan Effects of Muscle-Specific Androgen Receptor Overexpression on Body Composition of Male and Female Rats. Endocrinology 2024; 165:bqae012. [PMID: 38301268 DOI: 10.1210/endocr/bqae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 02/03/2024]
Abstract
Androgenic actions of gonadal testosterone are thought to be a major mechanism promoting sex differences in body composition across the lifespan. However, this inference is based on studies of androgen receptor (AR) function in late adolescent or emerging adult rodents. Here we assess body composition and AR expression in skeletal muscle of rats at defined ages, comparing wild-type (WT) to transgenic human skeletal actin-driven AR overexpression (HSAAR) rats which overexpress AR in skeletal muscle. Male and female HSAAR and WT Sprague Dawley rats (N = 288) underwent dual-energy x-ray absorptiometry (DXA) scanning and tissue collection at postnatal day (PND) 1, 10, 21, 42, 70, 183, 243, and 365. Expected sex differences in body composition and muscle mass largely onset with puberty (PND-21), with no associated changes to skeletal muscle AR protein. In adulthood, HSAAR increased tibialis anterior (TA) and extensor digitorum longus mass in males, and reduced the expected gain in gonadal fat mass in both sexes. In WT rats, AR protein was reduced in soleus, but not TA, throughout life. Nonetheless, soleus AR protein expression was greater in male rats than female rats at all ages of sexual development, yet only at PND-70 in TA. Overall, despite muscle AR overexpression effects, results are inconsistent with major sex differences in body composition during sexual development being driven by changes in muscle AR, rather suggesting that changes in ligand promote sexual differentiation of body composition during pubertal timing. Nonetheless, increased skeletal muscle AR in adulthood can be sufficient to increase muscle mass in males, and reduce adipose in both sexes.
Collapse
Affiliation(s)
- Sabrina Tzivia Barsky
- Department of Cell & Systems Biology, Faculty of Arts & Science, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Douglas Ashley Monks
- Department of Cell & Systems Biology, Faculty of Arts & Science, University of Toronto, Toronto, Ontario M5S 3G5, Canada
- Department of Psychology, Faculty of Arts & Science, University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada
| |
Collapse
|
2
|
Lacerda DR, Nunes-Silva A, Silveira ALM, Costa KA, Rodrigues DF, Moraes MM, Pinho V, Menezes GB, Teixeira MM, Wanner SP, Soares DD, Ferreira AVM. Acute exercise modulates the inflammatory response in adipose tissue in both lean and obese mice. Nutrition 2023; 115:112092. [PMID: 37549454 DOI: 10.1016/j.nut.2023.112092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/01/2023] [Accepted: 05/19/2023] [Indexed: 08/09/2023]
Abstract
OBJECTIVES Acute physical exercise acts as a metabolic stressor, promoting activation of the immune system, and this response could be relevant in the adipose tissue remodeling process. In addition, some cytokines have important functions in lipolysis. Because chronic exercise improves obesity-related metabolic and inflammatory dysfunction, herein we investigated the effect of acute exercise on the inflammatory responses in the adipose tissues of lean and obese mice. METHODS Lean mice were fed a standard chow diet, whereas obese mice were fed a high-refined carbohydrate diet for 8 wk. Both groups were subjected to 60 min of moderate-intensity exercise. RESULTS In the epididymal adipose tissue of lean mice, exercise enhanced interleukin (IL)-6 and tumor necrosis factor-α levels, which correlated positively with increased serum free fatty acid concentrations. In vivo confocal imaging of epididymal adipose tissue vessels revealed higher recruitment of neutrophils after exercise. Also, the number of leukocytes expressing CD11b+F480- was elevated 6 h after exercise. Similarly, the chemokine (C-X-C motif) ligand 1 level increased at 6 h and remained high until 24 h after exercise. Myeloperoxidase activity was increased at 6, 12, and 24 h after exercise. Surprisingly, however, no changes were observed in epididymal adipose tissue from obese mice, considering proinflammatory cytokines (IL-6 and tumor necrosis factor-α). On the other hand, IL-13, IL-4, and IL-10 levels were higher in obese mice after exercise. CONCLUSIONS These data suggest that acute exercise promotes an inflammatory response in the adipose tissue of lean mice that is observed as part of its role in adipose tissue remodeling. In contrast, acute exercise promotes an antiinflammatory response in adipose tissue from obese mice, likely as an important tool for restoring homeostasis.
Collapse
Affiliation(s)
- Débora Romualdo Lacerda
- Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Albená Nunes-Silva
- Department of Physical Education, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil.
| | | | - Kátia Anunciação Costa
- Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Débora Fernandes Rodrigues
- Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Michele Macedo Moraes
- Department of Physical Education, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vanessa Pinho
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gustavo Batista Menezes
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Samuel Penna Wanner
- Department of Physical Education, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Danusa Dias Soares
- Department of Physical Education, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | |
Collapse
|
3
|
Kim MW, Ham YJ, Kim HB, Lee JY, Lim JD, Lee HT. Anti-Obesity Effects of the Larval Powder of Steamed and Lyophilized Mature Silkworms in a Newly Designed Adult Mouse Model. Foods 2023; 12:3613. [PMID: 37835266 PMCID: PMC10572763 DOI: 10.3390/foods12193613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Recently, "mature" silkworms (MS) of Bombix mori have been considered a potential nutraceutical, with a number of health benefits reported for steamed and lyophilized MS powder (SMSP). However, no obesity-related effects have been reported for SMSP. In the present study, anti-obesity effects of SMSP were investigated in adult mice in vivo, aged 12 weeks at the onset of SMSP treatment, fed a normal diet (ND) and a high-fat diet (HFD), respectively, for 12 weeks. SMSP significantly suppressed body weight gain, intra-abdominal adipose tissue, and food efficiency in both ND-fed and HFD-fed adult mice. In addition, SMSP significantly decreased food intake and liver weight in HFD-fed mice, indicating that SMSP suppressed appetite and simultaneously reduced the conversion of feed into body weight in HFD-fed mice. SMSP also significantly lowered the serum levels of glucose, triglyceride, total cholesterol, low-density lipoprotein cholesterol, asparagine transaminase, alanine transaminase, and alkaline phosphatase. However, SMSP had no significant effect on the weights of the kidney, spleen, or thymus or the serum levels of blood urea nitrogen and creatinine. Taken together, the above results suggest that SMSP has potent anti-obesity effects and is safe for long-term use as a potential therapeutic and/or nutraceutical in both obese patients and non-obese individuals.
Collapse
Affiliation(s)
- Min Woo Kim
- Biopharmaceutical Engineering Major, Division of Applied Bioengineering, College of Engineering, Dong-eui University, Busan 47340, Republic of Korea; (M.W.K.); (Y.-J.H.)
| | - Yu-Jin Ham
- Biopharmaceutical Engineering Major, Division of Applied Bioengineering, College of Engineering, Dong-eui University, Busan 47340, Republic of Korea; (M.W.K.); (Y.-J.H.)
| | - Hyun-Bok Kim
- National Institute of Agricultural Sciences, Rural Development Administration, Wanju 55365, Republic of Korea;
| | - Ji young Lee
- Department of Ophthalmology and Visual Science, Daejeon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Jung-Dae Lim
- Department of Herbal Medicine Resource, Kangwon National University, Samcheok 25949, Republic of Korea;
| | - Hyun-Tai Lee
- Biopharmaceutical Engineering Major, Division of Applied Bioengineering, College of Engineering, Dong-eui University, Busan 47340, Republic of Korea; (M.W.K.); (Y.-J.H.)
- Core-Facility Center for Tissue Regeneration, Dong-Eui University, Busan 47340, Republic of Korea
| |
Collapse
|
4
|
Martinez ME, Wu Z, Hernandez A. Paternal developmental thyrotoxicosis disrupts neonatal leptin leading to increased adiposity and altered physiology of the melanocortin system. Front Endocrinol (Lausanne) 2023; 14:1210414. [PMID: 37560296 PMCID: PMC10407661 DOI: 10.3389/fendo.2023.1210414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 07/10/2023] [Indexed: 08/11/2023] Open
Abstract
Background The genetic code does not fully explain individual variability and inheritance of susceptibility to endocrine conditions, suggesting the contribution of epigenetic factors acting across generations. Methods We used a mouse model of developmental thyrotoxicosis (Dio3-/- mouse) to analyze endocrine outcomes in the adult offspring of Dio3-/- males using standard methods for body composition, and baseline and fasting hormonal and gene expression determinations in serum and tissues of relevance to the control of energy balance. Results Compared to controls, adult females with an exposed father (EF females) exhibited higher body weight and fat mass, but not lean mass, a phenotype that was much milder in EF males. After fasting, both EF females and males exhibited a more pronounced decrease in body weight than controls. EF females also showed markedly elevated serum leptin, increased white adipose tissue mRNA expression of leptin and mesoderm-specific transcript but decreased expression of type 2 deiodinase. EF females exhibited decreased serum ghrelin, which showed more pronounced post-fasting changes in EF females than in control females. EF female hypothalami also revealed significant decreases in the expression of pro-opiomelanocortin, agouti-related peptide, neuropeptide Y and melanocortin receptor 4. These markers also showed larger changes in response to fasting in EF females than in control females. Adult EF females showed no abnormalities in serum thyroid hormones, but pituitary expression of thyrotropin-releasing hormone receptor 1 and thyroid gland expression of thyroid-stimulating hormone receptor, thyroid peroxidase and iodotyrosine deiodinase were increased at baseline and showed differential regulation after fasting, with no increase in Trhr1 expression and more pronounced reductions in Tshr, Tpo and Iyd. In EF males, these abnormalities were generally milder. In addition, postnatal day 14 (P14) serum leptin was markedly reduced in EF pups. Discussion A paternal excess of thyroid hormone during development modifies the endocrine programming and energy balance in the offspring in a sexually dimorphic manner, with baseline and dynamic range alterations in the leptin-melanocortin system and thyroid gland, and consequences for adiposity phenotypes. We conclude that thyroid hormone overexposure may have important implications for the non-genetic, inherited etiology of endocrine and metabolic pathologies.
Collapse
Affiliation(s)
- Maria Elena Martinez
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, United States
| | - Zhaofei Wu
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, United States
| | - Arturo Hernandez
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, United States
- Graduate School for Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States
- Department of Medicine, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
5
|
Ferreira‐Baptista C, Queirós A, Ferreira R, Fernandes MH, Gomes PS, Colaço B. Retinoic acid induces the osteogenic differentiation of cat adipose tissue-derived stromal cells from distinct anatomical sites. J Anat 2023; 242:277-288. [PMID: 36056547 PMCID: PMC9877480 DOI: 10.1111/joa.13758] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/07/2022] [Accepted: 08/16/2022] [Indexed: 02/01/2023] Open
Abstract
Mesenchymal stromal cells-based regenerative orthopedic therapies have been used in cats as a promising and innovative therapeutic approach to enhance the repair of bone defects. Adipose tissue-derived stromal cells (ADSCs) can be obtained from two main sites-subcutaneous and visceral-with established differences regarding structure, composition, cell content, and functionality. However, in cats, to the best of the authors' knowledge, no studies have been conducted to compare the functional activity of the ADSCs isolated from the two sites, and the impact of these differences on the induced osteogenic potential. Additionally, retinoic acid has been recently regarded as a new osteogenic inducer within cells of distinct species, with undisclosed functionality on cat-derived cell populations. Thus, the present study aimed to evaluate the functional activity of ADSCs isolated from the subcutaneous and visceral adipose sites (SCAT and VAT, respectively) of the cat, as well as the effects of two osteogenic-inducing conditions-the classic dexamethasone, β-glycerophosphate and ascorbic acid-supplemented media (Dex + β + AAM), and Retinoic Acid-supplemented media (RAM). The adipose tissue of subcutaneous and visceral origin was isolated, characterized, and ADSCs were isolated and grown in the presence of the two osteogenic-inducing conditions, and characterized in terms of proliferation, metabolic activity, morphology, and osteogenic activity. Our results demonstrated a distinct biological profile of the two adipose tissue sites regarding cell size, vascularization, and morphology. Further, osteogenic-induced ADSCs from both sites presented an increased expression of alkaline phosphatase activity (ALP) and cytochemical staining, as compared with control. Overall, RAM induced higher levels of ALP activity than Dex + β + AAM, supporting an increased osteogenic activation. Additionally, VAT was the tissue with the best osteogenic potential, showing higher levels of ALP expression, particularly with RAM. In conclusion, different characteristics were found between the two adipose tissue sites-SCAT and VAT, which probably reflect the differences found in the functionality of isolated ADSCs from both tissues. Furthermore, for cat, VAT shows a greater osteogenic-inductive capacity than SCAT, particularly with RAM, which can be of therapeutic relevance for regenerative medicine applications.
Collapse
Affiliation(s)
- Carla Ferreira‐Baptista
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB)University of Trás‐os‐Montes e Alto Douro (UTAD)Vila RealPortugal
- BoneLab ‐ Laboratory for Bone Metabolism and Regeneration, Faculty of Dental MedicineUniversity of PortoPortoPortugal
- REQUIMTE/LAQVUniversity of PortoPortoPortugal
- REQUIMTE/LAQVDepartment of Chemistry University of AveiroAveiroPortugal
| | | | - Rita Ferreira
- REQUIMTE/LAQVDepartment of Chemistry University of AveiroAveiroPortugal
| | - Maria Helena Fernandes
- BoneLab ‐ Laboratory for Bone Metabolism and Regeneration, Faculty of Dental MedicineUniversity of PortoPortoPortugal
- REQUIMTE/LAQVUniversity of PortoPortoPortugal
| | - Pedro Sousa Gomes
- BoneLab ‐ Laboratory for Bone Metabolism and Regeneration, Faculty of Dental MedicineUniversity of PortoPortoPortugal
- REQUIMTE/LAQVUniversity of PortoPortoPortugal
| | - Bruno Colaço
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB)University of Trás‐os‐Montes e Alto Douro (UTAD)Vila RealPortugal
- REQUIMTE/LAQVUniversity of PortoPortoPortugal
- CECAV—Animal and Veterinary Research Centre UTADUniversity of Trás‐os‐Montes and Alto DouroVila RealPortugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS)Vila RealPortugal
| |
Collapse
|
6
|
The Osteogenic Potential of Falciform Ligament-Derived Stromal Cells-A Comparative Analysis between Two Osteogenic Induction Programs. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120810. [PMID: 36551016 PMCID: PMC9774535 DOI: 10.3390/bioengineering9120810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/28/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Mesenchymal stromal cells (MSCs) have gained special relevance in bone tissue regenerative applications. MSCs have been isolated from different depots, with adipose tissue being acknowledged as one of the most convenient sources, given the wide availability, high cellular yield, and obtainability. Recently, the falciform ligament (FL) has been regarded as a potential depot for adipose tissue-derived stromal cells (FL-ADSCs) isolation. Nonetheless, the osteogenic capability of FL-ADSCs has not been previously characterized. Thus, the present study aimed the detailed characterization of FL-ADSCs' functionality upon osteogenic induction through a classic (dexamethasone-based-DEX) or an innovative strategy with retinoic acid (RA) in a comparative approach with ADSCs from a control visceral region. Cultures were characterized for cell proliferation, metabolic activity, cellular morphology, fluorescent cytoskeletal and mitochondrial organization, and osteogenic activity-gene expression analysis and cytochemical staining. FL-derived populations expressed significantly higher levels of osteogenic genes and cytochemical markers, particularly with DEX induction, as compared to control ADSCs that were more responsive to RA. FL-ADSCs were identified as a potential source for bone regenerative applications, given the heightened osteogenic functionality. Furthermore, data highlighted the importance of the selection of the most adequate osteogenic-inducing program concerning the specificities of the basal cell population.
Collapse
|
7
|
Collins KH, Gui C, Ely EV, Lenz KL, Harris CA, Guilak F, Meyer GA. Leptin mediates the regulation of muscle mass and strength by adipose tissue. J Physiol 2022; 600:3795-3817. [PMID: 35844058 PMCID: PMC9378542 DOI: 10.1113/jp283034] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/04/2022] [Indexed: 02/01/2023] Open
Abstract
Adipose tissue secretes numerous cytokines (termed 'adipokines') that have known or hypothesized actions on skeletal muscle. The majority of adipokines have been implicated in the pathological link between excess adipose and muscle insulin resistance, but approximately half also have documented in vitro effects on myogenesis and/or hypertrophy. This complexity suggests a potential dual role for adipokines in the regulation of muscle mass in homeostasis and the development of pathology. In this study, we used lipodystrophic 'fat-free' mice to demonstrate that adipose tissue is indeed necessary for the development of normal muscle mass and strength. Fat-free mice had significantly reduced mass (∼15%) and peak contractile tension (∼20%) of fast-twitch muscles, a slowing of contractile dynamics and decreased cross-sectional area of fast twitch fibres compared to wild-type littermates. These deficits in mass and contractile tension were fully rescued by reconstitution of ∼10% of normal adipose mass, indicating that this phenotype is the direct consequence of absent adipose. We then showed that the rescue is solely mediated by the adipokine leptin, as similar reconstitution of adipose from leptin-knockout mice fails to rescue mass or strength. Together, these data indicate that the development of muscle mass and strength in wild-type mice is dependent on adipose-secreted leptin. This finding extends our current understanding of the multiple roles of adipokines in physiology as well as disease pathophysiology to include a critical role for the adipokine leptin in muscle homeostasis. KEY POINTS: Adipose-derived cytokines (adipokines) have long been implicated in the pathogenesis of insulin resistance in obesity but likely have other under-appreciated roles in muscle physiology. Here we use a fat-free mouse to show that adipose tissue is necessary for the normal development of muscle mass and strength. Through add-back of genetically modified adipose tissue we show that leptin is the key adipokine mediating this regulation. This expands our understanding of leptin's role in adipose-muscle signalling to include development and homeostasis and adds the surprising finding that leptin is the sole mediator of the maintenance of muscle mass and strength by adipose tissue.
Collapse
Affiliation(s)
- Kelsey H. Collins
- Department of Orthopaedic SurgeryWashington University in St. LouisMOUSA,Shriners Hospitals for ChildrenSt LouisMOUSA,Center of Regenerative MedicineWashington University in St. LouisMOUSA
| | - Chang Gui
- Department of Biomedical EngineeringWashington University in St. LouisMOUSA,Program in Physical TherapyWashington UniversitySt LouisMOUSA
| | - Erica V. Ely
- Department of Orthopaedic SurgeryWashington University in St. LouisMOUSA,Shriners Hospitals for ChildrenSt LouisMOUSA,Center of Regenerative MedicineWashington University in St. LouisMOUSA,Department of Biomedical EngineeringWashington University in St. LouisMOUSA
| | - Kristin L. Lenz
- Department of Orthopaedic SurgeryWashington University in St. LouisMOUSA,Shriners Hospitals for ChildrenSt LouisMOUSA,Center of Regenerative MedicineWashington University in St. LouisMOUSA
| | - Charles A. Harris
- Division of EndocrinologyMetabolism & Lipid ResearchWashington UniversitySt LouisMissouriUSA
| | - Farshid Guilak
- Department of Orthopaedic SurgeryWashington University in St. LouisMOUSA,Shriners Hospitals for ChildrenSt LouisMOUSA,Center of Regenerative MedicineWashington University in St. LouisMOUSA,Department of Biomedical EngineeringWashington University in St. LouisMOUSA
| | - Gretchen A. Meyer
- Department of Orthopaedic SurgeryWashington University in St. LouisMOUSA,Center of Regenerative MedicineWashington University in St. LouisMOUSA,Department of Biomedical EngineeringWashington University in St. LouisMOUSA,Program in Physical TherapyWashington UniversitySt LouisMOUSA,Department of NeurologyWashington University in St. LouisSt LouisMOUSA
| |
Collapse
|
8
|
Aiassa V, Del Rosario Ferreira M, Villafañe N, Eugenia D'Alessandro M. α-Linolenic acid rich-chia seed modulates visceral adipose tissue collagen deposition, lipolytic enzymes expression, insulin signaling and GLUT-4 levels in a diet-induced adiposity rodent model. Food Res Int 2022; 156:111164. [PMID: 35651030 DOI: 10.1016/j.foodres.2022.111164] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/28/2022] [Accepted: 03/15/2022] [Indexed: 11/19/2022]
Abstract
Given obesity and its associated metabolic disorders have reached epidemic proportions, the study of therapeutic strategies targeting white adipose tissue (WAT) are of main research interest. We previously shown that α-linolenic acid-rich chia seed was able to ameliorate a wide range of metabolic disorders including body fat accretion in sucrose-rich diet (SRD)-fed rats, an experimental model of visceral adiposity and insulin resistance. However, the mechanisms involved are not fully clarified. The aim of this study was to evaluate the effect of chia seed administration upon WAT remodeling and key enzymes that controls lipolysis, insulin signaling (tAKT, pAKT), and GLUT-4 levels in different visceral fat pad depots (epididymal -eWAT- and retroperitoneal -rWAT- adipose tissues) of SRD-fed rats. Results showed that chia seed reduces adipocytes hypertrophy, the increased lipid content and collagen deposition in both WAT. These changes were accompanied by a significant reduction of HSL and ATGL protein levels in eWAT and HSL protein levels in rWAT. Moreover, chia seed restored the altered expression pattern of the pAKT observed in SRD-fed rats, and modulated GLUT-4 levels. Chia seed could be a dietary intervention of great relevance with potential beneficial effects in the management of body fat excess and WAT function.
Collapse
Affiliation(s)
- Victoria Aiassa
- Laboratorio de Estudio de Enfermedades Metabólicas relacionadas con la Nutrición. Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, Santa Fe, Argentina
| | - María Del Rosario Ferreira
- Laboratorio de Estudio de Enfermedades Metabólicas relacionadas con la Nutrición. Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, Santa Fe, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Noelia Villafañe
- Departamento de Morfología. Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, Santa Fe, Argentina
| | - María Eugenia D'Alessandro
- Laboratorio de Estudio de Enfermedades Metabólicas relacionadas con la Nutrición. Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, Santa Fe, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina.
| |
Collapse
|
9
|
Secreted Protein Acidic and Rich in Cysteine (Sparc) KO Leads to an Accelerated Ageing Phenotype Which Is Improved by Exercise Whereas SPARC Overexpression Mimics Exercise Effects in Mice. Metabolites 2022; 12:metabo12020125. [PMID: 35208200 PMCID: PMC8879002 DOI: 10.3390/metabo12020125] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/18/2022] [Accepted: 01/24/2022] [Indexed: 01/04/2023] Open
Abstract
Secreted protein acidic and rich in cysteine (SPARC) is a matricellular glycoprotein implicated in various functions, including metabolism, tissue regeneration, and functional homeostasis. SPARC/Sparc declines with ageing but increases with exercise. We aim to verify two hypotheses: (1) SPARC deficiency leads to an ageing-like phenotype (metabolic decline, muscle loss, etc.), and (2) SPARC overexpression would mimic exercise, counteract ageing, and improve age-related changes. Our mice experiments are divided into two parts. First, we explore the consequences of Sparc knockout (KO) and compare them to the ageing effects. We also observe the effects of exercise. In the second part, we study the effects of SPARC overexpression and compare them to the exercise benefits. At the end, we make an analysis of the results to point out the analogies between Sparc KO and the ageing-like phenotype on the one hand and make comparisons between SPARC overexpression and exercise in the context of exercise counteracting ageing. The measurements were mainly related to tissue weights, adiposity, metabolism, and muscle strength. The main findings are that Sparc KO reduced glucose tolerance, muscle glucose transporter expression, and abdominal adipose tissue weight but increased glycogen content in the muscle. SPARC overexpression increased muscle strength, muscle mass, and expressions of the muscle glucose transporter and mitochondrial oxidative phosphorylation but lowered the glycemia and the adiposity, especially in males. Collectively, these findings, and the data we have previously reported, show that Sparc KO mice manifest an ageing-like phenotype, whereas SPARC overexpression and exercise generate similar benefits. The benefits are towards counteracting both the SPARC deficiency-induced ageing-like phenotype as well as reversing the age-related changes. The potential applications of these findings are to build/optimize Sparc KO-based animal models of various health conditions and, on the other hand, to develop therapies based on introducing SPARC or targeting SPARC-related pathways to mimic exercise against age-related and metabolic disorders.
Collapse
|
10
|
Chun KH. Mouse model of the adipose organ: the heterogeneous anatomical characteristics. Arch Pharm Res 2021; 44:857-875. [PMID: 34606058 DOI: 10.1007/s12272-021-01350-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/20/2021] [Indexed: 12/24/2022]
Abstract
Adipose tissue plays a pivotal role in energy storage, hormone secretion, and temperature control. Mammalian adipose tissue is largely divided into white adipose tissue and brown adipose tissue, although recent studies have discovered the existence of beige adipocytes. Adipose tissues are widespread over the whole body and each location shows distinctive metabolic features. Mice are used as a representative experimental model system in metabolic studies due to their numerous advantages. Importantly, the adipose tissues of experimental animals and humans are not perfectly matched, and each adipose tissue exhibits both similar and specific characteristics. Nevertheless, the diversity and characteristics of mouse adipose tissue have not yet been comprehensively summarized. This review summarizes diverse information about the different types of adipose tissue being studied in mouse models. The types and characteristics of adipocytes were described, and each adipose tissue was classified by type, and features such as its distribution, origin, differences from humans, and metabolic characteristics were described. In particular, the distribution of widely studied adipose tissues was illustrated so that researchers can comprehensively grasp its location. Also, the adipose tissues misused or confusingly used among researchers were described. This review will provide researchers with comprehensive information and cautions needed to study adipose tissues in mouse models.
Collapse
Affiliation(s)
- Kwang-Hoon Chun
- Gachon Institute of Pharmaceutical Sciences, College of Pharmacy, Gachon University, Inchon, 21936, Republic of Korea.
| |
Collapse
|
11
|
Clemente-Olivo MP, Habibe JJ, Vos M, Ottenhoff R, Jongejan A, Herrema H, Zelcer N, Kooijman S, Rensen PCN, van Raalte DH, Nieuwdorp M, Eringa EC, de Vries CJ. Four-and-a-half LIM domain protein 2 (FHL2) deficiency protects mice from diet-induced obesity and high FHL2 expression marks human obesity. Metabolism 2021; 121:154815. [PMID: 34119536 DOI: 10.1016/j.metabol.2021.154815] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/26/2021] [Accepted: 06/08/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Four-and-a-Half-LIM-domain-protein 2 (FHL2) modulates multiple signal transduction pathways but has not been implicated in obesity or energy metabolism. In humans, methylation and expression of the FHL2 gene increases with age, and high FHL2 expression is associated with increased body weight in humans and mice. This led us to hypothesize that FHL2 is a determinant of diet-induced obesity. METHODS FHL2-deficient (FHL2-/-) and wild type male mice were fed a high-fat diet. Metabolic phenotyping of these mice, as well as transcriptional analysis of key metabolic tissues was performed. Correlation of the expression of FHL2 and relevant genes was assessed in datasets from white adipose tissue of individuals with and without obesity. RESULTS FHL2 Deficiency protects mice from high-fat diet-induced weight gain, whereas glucose handling is normal. We observed enhanced energy expenditure, which may be explained by a combination of changes in multiple tissues; mild activation of brown adipose tissue with increased fatty acid uptake, increased cardiac glucose uptake and browning of white adipose tissue. Corroborating our findings in mice, expression of FHL2 in human white adipose tissue positively correlates with obesity and negatively with expression of browning-associated genes. CONCLUSION Our results position FHL2 as a novel regulator of obesity and energy expenditure in mice and human. Given that FHL2 expression increases during aging, we now show that low FHL2 expression associates with a healthy metabolic state.
Collapse
Affiliation(s)
- Maria P Clemente-Olivo
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Jayron J Habibe
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands; Department of Physiology, Amsterdam UMC, Amsterdam Cardiovascular Sciences, location VUmc, Amsterdam, the Netherlands
| | - Mariska Vos
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Roelof Ottenhoff
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Aldo Jongejan
- Department of Bioinformatics, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Hilde Herrema
- Department of Experimental Vascular Medicine, Amsterdam UMC, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Noam Zelcer
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Sander Kooijman
- Department of Medicine, Division of Endocrinology, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Daniël H van Raalte
- Department of Internal Medicine, Diabetes Center, Amsterdam UMC, Amsterdam Cardiovascular Sciences Amsterdam, the Netherlands
| | - Max Nieuwdorp
- Department of Experimental Vascular Medicine, Amsterdam UMC, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Etto C Eringa
- Department of Physiology, Amsterdam UMC, Amsterdam Cardiovascular Sciences, location VUmc, Amsterdam, the Netherlands
| | - Carlie J de Vries
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands.
| |
Collapse
|
12
|
Chen R, Huang S, Lin T, Ma H, Shan W, Duan F, Lv J, Zhang J, Ren L, Nie L. Photoacoustic molecular imaging-escorted adipose photodynamic-browning synergy for fighting obesity with virus-like complexes. NATURE NANOTECHNOLOGY 2021; 16:455-465. [PMID: 33526836 DOI: 10.1038/s41565-020-00844-6] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 12/17/2020] [Indexed: 06/12/2023]
Abstract
Photodynamic therapy and adipose browning induction are two promising approaches to reverse obesity. The former strategy acts rapidly and locally, whereas the latter has a more gradual and widespread effect. Despite their complementarity, they have rarely been combined and imaged non-invasively in vivo. Here we introduce an adipose-targeting hepatitis B core protein complex that contains a traceable photosensitizer (ZnPcS4 (zinc phthalocyanine tetrasulfonate)) and a browning agent (rosiglitazone) that allows simultaneous photodynamic and browning treatments, with photoacoustic molecular imaging. After intravenous injection in obese mice, the complex binds specifically to white adipose tissues, especially those rich in blood supply, and drives adipose reduction thanks to the synergy of ZnPcS4 photodynamics and rosiglitazone browning. Using photoacoustic molecular imaging, we could monitor the changes induced by the treatment, which included complex activity, lipid catabolism and angiogenesis. Our findings demonstrate the anti-obesity potential of our feedback-based synergic regimen orchestrated by the targeted hepatitis B core complex.
Collapse
Affiliation(s)
- Ronghe Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnosis & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Shanshan Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnosis & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Tongtong Lin
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surface, College of Materials, Xiamen University, Xiamen, China
| | - Haosong Ma
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnosis & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Wenjun Shan
- Department of Pharmacology, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, China
| | - Fei Duan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnosis & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Jing Lv
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnosis & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Jinde Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnosis & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Lei Ren
- Department of Biomaterials, Key Laboratory of Biomedical Engineering of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surface, College of Materials, Xiamen University, Xiamen, China
| | - Liming Nie
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnosis & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China.
| |
Collapse
|
13
|
Eshima H, Tamura Y, Kakehi S, Kakigi R, Hashimoto R, Funai K, Kawamori R, Watada H. A chronic high-fat diet exacerbates contractile dysfunction with impaired intracellular Ca 2+ release capacity in the skeletal muscle of aged mice. J Appl Physiol (1985) 2020; 128:1153-1162. [PMID: 32213111 DOI: 10.1152/japplphysiol.00530.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Obesity and aging reduce skeletal muscle contractile function. However, it remains unclear whether obesity additively promotes muscle contractile dysfunction in the setting of aging. In this study, we investigated skeletal muscle contractile function ex vivo and intracellular Ca2+ release in male C57BL/6J mice fed a low-fat diet (LFD) or a high-fat diet (HFD) for 4 or 20 mo. Tetanic force production in the extensor digitorum longus muscle was decreased by aging or HFD feeding, and the further reduction was observed in aged HFD mice. The 20-mo HFD-fed mice, not the 20-mo LFD-fed mice or 4-mo HFD-fed mice, showed reduced intracellular Ca2+ peak levels by high concentration of caffeine (25 mM) compared with 4-mo LFD mice. Aging and HFD feeding additively increased intramyocellular lipid (IMCL) levels and were associated with the degree of impaired muscle contractile force and peak Ca2+ level. These data suggest that impairment in the contractile force in aged muscle is aggravated by HFD, which may be due, at least in part, to dysfunction in intracellular Ca2+ release. The IMCL level may be a marker for impaired muscle contractile force caused by aging and HFD.NEW & NOTEWORTHY The aim of this study was to examine the effect of high-fat diet (HFD)-induced obesity on contractile function and Ca2+ release capacity in aged skeletal muscle. Not only were the force production and peak Ca2+ levels decreased by aging and HFD feeding, respectively, but also, these interventions had an additive effect in aged HFD-fed mice. These data suggest that the impairment in the contractile force in aged muscle is aggravated by a HFD, which may be due to synergistic dysfunction in intracellular Ca2+ release.
Collapse
Affiliation(s)
- Hiroaki Eshima
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, Utah
| | - Yoshifumi Tamura
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Saori Kakehi
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ryo Kakigi
- Department of Physiology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Ryota Hashimoto
- Department of Physiology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Katsuhiko Funai
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, Utah
| | - Ryuzo Kawamori
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hirotaka Watada
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Center for Therapeutic Innovations in Diabetes, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Center for Molecular Diabetology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
14
|
Val CH, de Oliveira MC, Lacerda DR, Barroso A, Batista NV, Menezes-Garcia Z, de Assis DRR, Cramer AT, Brant F, Teixeira MM, Glória Souza D, Ferreira AM, Machado FS. SOCS2 modulates adipose tissue inflammation and expansion in mice. J Nutr Biochem 2019; 76:108304. [PMID: 31816561 DOI: 10.1016/j.jnutbio.2019.108304] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 11/06/2019] [Accepted: 11/12/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Obesity is usually triggered by a nutrient overload that favors adipocyte hypertrophy and increases the number of pro-inflammatory cells and mediators into adipose tissue. These mediators may be regulated by suppressors of cytokine signaling (SOCS), such as SOCS2, which is involved in the regulation of the inflammatory response of many diseases, but its role in obesity is not yet known. We aimed to investigate the role of SOCS2 in metabolic and inflammatory dysfunction induced by a high-refined carbohydrate-containing diet (HC). MATERIAL AND METHODS Male C57BL/6 wild type (WT) and SOCS2 deficient (SOCS2-/-) mice were fed chow or an HC diet for 8 weeks. RESULTS In general, SOCS2 deficient mice, independent of the diet, showed higher adipose tissue mass compared with their WT counterparts that were associated with decreased lipogenesis rate in adipose tissue, lipolysis in adipocyte culture and energy expenditure. An anti-inflammatory profile was observed in adipose tissue of SOCS2-/- by reduced secretion of cytokines, such as TNF and IL-6, and increased M2-like macrophages and regulatory T cells compared with WT mice. Also, SOCS2 deficiency reduced the differentiation/expansion of pro-inflammatory cells in the spleen but increased Th2 and Treg cells compared with their WT counterparts. CONCLUSION The SOCS2 protein is an important modulator of obesity that regulates the metabolic pathways related to adipocyte size. Additionally, SOCS2 is an inflammatory regulator that appears to be essential for controlling the release of cytokines and the differentiation/recruitment of cells into adipose tissue during the development of obesity.
Collapse
Affiliation(s)
- Cynthia Honorato Val
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Brazil
| | | | | | - Andreia Barroso
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Brazil
| | | | | | | | | | - Fátima Brant
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Brazil
| | | | | | | | - Fabiana Simão Machado
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Brazil.
| |
Collapse
|
15
|
Takei R, Inoue T, Sonoda N, Kohjima M, Okamoto M, Sakamoto R, Inoguchi T, Ogawa Y. Bilirubin reduces visceral obesity and insulin resistance by suppression of inflammatory cytokines. PLoS One 2019; 14:e0223302. [PMID: 31577826 PMCID: PMC6774504 DOI: 10.1371/journal.pone.0223302] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/18/2019] [Indexed: 02/08/2023] Open
Abstract
Objective Although previous studies have reported a negative relationship between serum bilirubin concentration and the development of diabetes mellitus (DM), the relationship between bilirubin and insulin resistance has not been thoroughly assessed. This study was designed to determine the relationships between bilirubin, body fat distribution, and adipose tissue inflammation in patients with type 2 DM and the effect of bilirubin in an obese animal model. Method Body fat distribution was measured using an abdominal dual bioelectrical impedance analyzer in patients with type 2 DM. We also measured glycemic control, lipid profile, serum bilirubin concentration and other clinical characteristics, and determined their relationships with body fat distribution. In the animal study, biliverdin (20 mg/kg daily) was orally administered to high-fat diet (HFD)-induced obese (DIO) mice for 2 weeks, after which intraperitoneal insulin tolerance testing was performed. Then, adipocyte area, adipocytokine expression, and macrophage polarization were evaluated in epididymal adipose tissues. Results In the clinical study, univariate analysis showed that a lower bilirubin concentration was significantly correlated with higher body mass index, waist circumference, triglyceride, uric acid, creatinine, visceral fat area and lower HDL-C. In multivariate analyses, bilirubin concentration significantly correlated with diastolic blood pressure, creatinine, and visceral fat area. However, there was no association between bilirubin concentration and subcutaneous fat area. In the animal study, DIO mice treated with biliverdin had smaller adipocytes than untreated DIO mice and biliverdin improved HFD-induced insulin resistance. Biliverdin treatment reversed the higher gene expression of Cd11c, encoding an M1 macrophage marker, and Tnfa, encoding the proinflammatory cytokine tumor necrosis factor-α, in the adipose tissues of DIO mice. These data suggest biliverdin administration alleviates insulin resistance by ameliorating inflammation and the dysregulation of adipocytokine expression in adipose tissues of DIO mice. Conclusions Bilirubin may protect against insulin resistance by ameliorating visceral obesity and adipose tissue inflammation.
Collapse
Affiliation(s)
- Ryoko Takei
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Fukuoka City Health Promotion Support Center, Fukuoka, Japan
| | - Tomoaki Inoue
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Noriyuki Sonoda
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Motoyuki Kohjima
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Misato Okamoto
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryuichi Sakamoto
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toyoshi Inoguchi
- Fukuoka City Health Promotion Support Center, Fukuoka, Japan
- * E-mail:
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Molecular and Cellular Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
16
|
Transcriptional profiling identifies strain-specific effects of caloric restriction and opposite responses in human and mouse white adipose tissue. Aging (Albany NY) 2019; 10:701-746. [PMID: 29708498 PMCID: PMC5940131 DOI: 10.18632/aging.101424] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 04/20/2018] [Indexed: 12/13/2022]
Abstract
Caloric restriction (CR) has been extensively studied in rodents as an intervention to improve lifespan and healthspan. However, effects of CR can be strain- and species-specific. This study used publically available microarray data to analyze expression responses to CR in males from 7 mouse strains (C57BL/6J, BALB/c, C3H, 129, CBA, DBA, B6C3F1) and 4 tissues (epididymal white adipose tissue (eWAT), muscle, heart, cortex). In each tissue, the largest number of strain-specific CR responses was identified with respect to the C57BL/6 strain. In heart and cortex, CR responses in C57BL/6 mice were negatively correlated with responses in other strains. Strain-specific CR responses involved genes associated with olfactory receptors (Olfr1184, Olfr910) and insulin/IGF-1 signaling (Igf1, Irs2). In each strain, CR responses in eWAT were negatively correlated with those in human subcutaneous WAT (scWAT). In human scWAT, CR increased expression of genes associated with stem cell maintenance and vascularization. However, orthologous genes linked to these processes were down-regulated in mouse. These results identify strain-specific CR responses limiting generalization across mouse strains. Differential CR responses in mouse versus human WAT may be due to differences in the depots examined and/or the presence of “thrifty genes” in humans that resist adipose breakdown despite caloric deficit.
Collapse
|
17
|
López-Fontana CM, Pennacchio G, Zyla LE, Toneatto J, Bruna FA, Ortiz N, Sassi PL, Santiano FE, García S, Sasso CV, Pietrobon EO, Jahn GA, Pistone Creydt V, Soaje M, Carón RW. Effects of hypothyroidism on the mesenteric and omental adipose tissue in rats. Mol Cell Endocrinol 2019; 490:88-99. [PMID: 31004687 DOI: 10.1016/j.mce.2019.04.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 04/14/2019] [Accepted: 04/15/2019] [Indexed: 12/27/2022]
Abstract
To characterize the influence of hypothyroidism on the endocrine activity of mesenteric and omental adipose tissue (MOAT) and the peripheral regulation of energy balance (EB) in rats, we analyzed food intake (FI); basal metabolic rate (BMR); locomotor activity; body weight (BW); serum hormone concentrations and the expression of their receptors in MOAT. We evaluated the morphology and differentiation of adipocytes. Hypothyroidism decreased FI, BMR and BW. The percentage of visceral white adipose tissue (WAT) depots and the morphology of adipocytes were similar to euthyroid rats. Serum leptin and adiponectin expression in MOAT were altered by hypothyroidism. The expression of Perilipin 1, HSL, UCP1 and PRDM16 was significantly lower in MOAT of hypothyroid animals. Hypothyroidism in rats leads to a compensated EB by inducing a white adipocyte dysfunction and a decrease in BW, BMR, FI and adipokine secretions without changing the percentage of WAT depots and the morphology of the MOAT.
Collapse
Affiliation(s)
- C M López-Fontana
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, CCT-Mendoza, Argentina.
| | - G Pennacchio
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, CCT-Mendoza, Argentina.
| | - L E Zyla
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, CCT-Mendoza, Argentina.
| | - J Toneatto
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina.
| | - F A Bruna
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, CCT-Mendoza, Argentina.
| | - N Ortiz
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, CCT-Mendoza, Argentina.
| | - P L Sassi
- Instituto Argentino de Investigaciones de las Zonas Áridas (IADIZA), CONICET, CCT-Mendoza, Argentina.
| | - F E Santiano
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, CCT-Mendoza, Argentina.
| | - S García
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, CCT-Mendoza, Argentina.
| | - C V Sasso
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, CCT-Mendoza, Argentina.
| | - E O Pietrobon
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, CCT-Mendoza, Argentina.
| | - G A Jahn
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, CCT-Mendoza, Argentina.
| | - V Pistone Creydt
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, CCT-Mendoza, Argentina.
| | - M Soaje
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, CCT-Mendoza, Argentina.
| | - R W Carón
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, CCT-Mendoza, Argentina.
| |
Collapse
|
18
|
Evans BA, Merlin J, Bengtsson T, Hutchinson DS. Adrenoceptors in white, brown, and brite adipocytes. Br J Pharmacol 2019; 176:2416-2432. [PMID: 30801689 DOI: 10.1111/bph.14631] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/28/2019] [Accepted: 02/11/2019] [Indexed: 01/01/2023] Open
Abstract
Adrenoceptors play an important role in adipose tissue biology and physiology that includes regulating the synthesis and storage of triglycerides (lipogenesis), the breakdown of stored triglycerides (lipolysis), thermogenesis (heat production), glucose metabolism, and the secretion of adipocyte-derived hormones that can control whole-body energy homeostasis. These processes are regulated by the sympathetic nervous system through actions at different adrenoceptor subtypes expressed in adipose tissue depots. In this review, we have highlighted the role of adrenoceptor subtypes in white, brown, and brite adipocytes in both rodents and humans and have included detailed analysis of adrenoceptor expression in human adipose tissue and clonally derived adipocytes. We discuss important considerations when investigating adrenoceptor function in adipose tissue or adipocytes. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.
Collapse
Affiliation(s)
- Bronwyn A Evans
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Jon Merlin
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Tore Bengtsson
- Department of Molecular Biosciences, The Wenner-Gren Institute, The Arrhenius Laboratories F3, Stockholm University, Stockholm, Sweden
| | - Dana S Hutchinson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| |
Collapse
|
19
|
McMurphy T, Huang W, Liu X, Siu JJ, Queen NJ, Xiao R, Cao L. Hypothalamic gene transfer of BDNF promotes healthy aging in mice. Aging Cell 2019; 18:e12846. [PMID: 30585393 PMCID: PMC6413658 DOI: 10.1111/acel.12846] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/15/2018] [Accepted: 09/02/2018] [Indexed: 12/26/2022] Open
Abstract
The aging process and age-related diseases all involve perturbed energy adaption and impaired ability to cope with adversity. Brain-derived neurotrophic factor (BDNF) in the hypothalamus plays important role in regulation of energy balance. Our previous studies show that recombinant adeno-associated virus (AAV)-mediated hypothalamic BDNF gene transfer alleviates obesity, diabetes, and metabolic syndromes in both diet-induced and genetic models. Here we examined the efficacy and safety of a built-in autoregulatory system to control transgene BDNF expression mimicking the body's natural feedback systems in middle-aged mice. Twelve-month-old mice were treated with either autoregulatory BDNF vector or yellow fluorescence protein (YFP) control, maintained on normal diet, and monitored for 28 weeks. BDNF gene transfer prevented the development of aging-associated metabolic declines characterized by: preventing aging-associated weight gain, reducing adiposity, reversing the decline of brown fat activity, increasing adiponectin while reducing leptin and insulin in circulation, improving glucose tolerance, increasing energy expenditure, alleviating hepatic steatosis, and suppressing inflammatory genes in the hypothalamus and adipose tissues. Moreover, BDNF treatment reduced anxiety-like and depression-like behaviors. These safety and efficacy data provide evidence that hypothalamic BDNF is a target for promoting healthy aging.
Collapse
Affiliation(s)
- Travis McMurphy
- Department of Cancer Biology and Genetics, College of MedicineThe Ohio State UniversityColumbusOhio
- The Ohio State University Comprehensive Cancer CenterColumbusOhio
| | - Wei Huang
- Department of Cancer Biology and Genetics, College of MedicineThe Ohio State UniversityColumbusOhio
- The Ohio State University Comprehensive Cancer CenterColumbusOhio
| | - Xianglan Liu
- Department of Cancer Biology and Genetics, College of MedicineThe Ohio State UniversityColumbusOhio
- The Ohio State University Comprehensive Cancer CenterColumbusOhio
| | - Jason J. Siu
- Department of Cancer Biology and Genetics, College of MedicineThe Ohio State UniversityColumbusOhio
- The Ohio State University Comprehensive Cancer CenterColumbusOhio
| | - Nicholas J. Queen
- Department of Cancer Biology and Genetics, College of MedicineThe Ohio State UniversityColumbusOhio
- The Ohio State University Comprehensive Cancer CenterColumbusOhio
| | - Run Xiao
- Department of Cancer Biology and Genetics, College of MedicineThe Ohio State UniversityColumbusOhio
- The Ohio State University Comprehensive Cancer CenterColumbusOhio
| | - Lei Cao
- Department of Cancer Biology and Genetics, College of MedicineThe Ohio State UniversityColumbusOhio
- The Ohio State University Comprehensive Cancer CenterColumbusOhio
| |
Collapse
|
20
|
Li X, Wang Z, Klaunig JE. The effects of perfluorooctanoate on high fat diet induced non-alcoholic fatty liver disease in mice. Toxicology 2019; 416:1-14. [DOI: 10.1016/j.tox.2019.01.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/21/2019] [Accepted: 01/29/2019] [Indexed: 01/28/2023]
|
21
|
Bodur A, İnce İ, Kahraman C, Abidin İ, Aydin-Abidin S, Alver A. Effect of a high sucrose and high fat diet in BDNF (+/-) mice on oxidative stress markers in adipose tissues. Arch Biochem Biophys 2019; 665:46-56. [PMID: 30797748 DOI: 10.1016/j.abb.2019.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 12/12/2022]
Abstract
The purpose of this study was to investigate the effects of a high fat and a high sucrosediet in wild type and BDNF (+/-) mice on oxidative stress in epididymal and subcutaneousadipose tissues by measuring different markers of oxidative stress and antioxidant enzymes. Wild type (WT) and BDNF (+/-) male mice were divided into six groups receiving fed control diet (CD), high sucrose diet (HSD), or high fat diet (HFD) for four months. Levels of 3-nitrotyrosine (3-NT) increased in the HFD-fed BDNF (+/-) mice, while 4-hydroxynonenal (4-HNE) levels increased in the CD and HFD-fed BDNF (+/-) groups. Malondialdehyde (MDA) levels decreased in subcutaneous tissue compared to epididymal adipose tissue, independently of diet type. Superoxide dismutase (SOD) activity was reduced by HFD (p < 0.05), butglutathione peroxidase (GSH-Px) activity was increased by HSD in epididymal adipose tissuein BDNF (+/-) mice (p < 0.05). GSH-Px activities was increased by CD and HFD in subcutaneous adipose tissue of BDNF (+/-) (p < 0.05). SOD2 and GSH-Px3 expressions were only decreased by HSD in epididymal and subcutaneous adipose tissues of BDNF (+/-) mice (p < 0.05). In conclusion, reduced BDNF may increase OS in epididymal adipose tissue, but not in subcutaneous adipose tissue following HSD and HFD.
Collapse
Affiliation(s)
- Akın Bodur
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - İmran İnce
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Cemil Kahraman
- Department of Nutrition and Dietetics, School of Health, Düzce University, Düzce, Turkey
| | - İsmail Abidin
- Department of Biophysics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Selcen Aydin-Abidin
- Department of Biophysics, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Ahmet Alver
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey.
| |
Collapse
|
22
|
Illesca P, Valenzuela R, Espinosa A, Echeverría F, Soto-Alarcon S, Ortiz M, Videla LA. Hydroxytyrosol supplementation ameliorates the metabolic disturbances in white adipose tissue from mice fed a high-fat diet through recovery of transcription factors Nrf2, SREBP-1c, PPAR-γ and NF-κB. Biomed Pharmacother 2019; 109:2472-2481. [DOI: 10.1016/j.biopha.2018.11.120] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/19/2018] [Accepted: 11/25/2018] [Indexed: 12/26/2022] Open
|
23
|
Leclerc D, Christensen KE, Cauvi O, Yang E, Fournelle F, Bahous RH, Malysheva OV, Deng L, Wu Q, Zhou Z, Gao ZH, Chaurand P, Caudill MA, Rozen R. Mild Methylenetetrahydrofolate Reductase Deficiency Alters Inflammatory and Lipid Pathways in Liver. Mol Nutr Food Res 2018; 63:e1801001. [PMID: 30408316 DOI: 10.1002/mnfr.201801001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/29/2018] [Indexed: 12/13/2022]
Abstract
SCOPE Dietary and genetic folate disturbances can lead to nonalcoholic fatty liver disease (NAFLD). A common variant in methylenetetrahydrofolate reductase (MTHFR 677C→T) causes mild MTHFR deficiency with lower 5-methyltetrahydrofolate for methylation reactions. The goal is to determine whether mild murine MTHFR deficiency contributes to NAFLD-related effects. METHODS AND RESULTS Wild-type and Mthfr+/- mice, a model for the human variant, are fed control (CD) or high-fat (HFAT) diets for 8 weeks. On both diets, MTHFR deficiency results in decreased S-adenosylmethionine, increased S-adenosylhomocysteine, and decreased betaine with reduced methylation capacity, and changes in expression of several inflammatory or anti-inflammatory mediators (Saa1, Apoa1, and Pon1). On CD, MTHFR deficiency leads to microvesicular steatosis with expression changes in lipid regulators Xbp1s and Cyp7a1. The combination of MTHFR deficiency and HFAT exacerbates changes in inflammatory mediators and introduces additional effects on inflammation (Saa2) and lipid metabolism (Nr1h4, Srebf1c, Ppara, and Crot). These effects are consistent with increased expression of pro-inflammatory HDL precursors and greater lipid accumulation. MTHFR deficiency may enhance liver injury through alterations in methylation capacity, inflammatory response, and lipid metabolism. CONCLUSION Individuals with the MTHFR variant may be at increased risk for liver disease and related complications, particularly when consuming high-fat diets.
Collapse
Affiliation(s)
- Daniel Leclerc
- Departments of Human Genetics and Pediatrics, McGill University, McGill University Health Center (MUHC), Montreal, H4A 3J1, Canada
| | - Karen E Christensen
- Departments of Human Genetics and Pediatrics, McGill University, McGill University Health Center (MUHC), Montreal, H4A 3J1, Canada
| | - Olivia Cauvi
- Departments of Human Genetics and Pediatrics, McGill University, McGill University Health Center (MUHC), Montreal, H4A 3J1, Canada
| | - Ethan Yang
- Department of Chemistry, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Frédéric Fournelle
- Department of Chemistry, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Renata H Bahous
- Departments of Human Genetics and Pediatrics, McGill University, McGill University Health Center (MUHC), Montreal, H4A 3J1, Canada
| | - Olga V Malysheva
- Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY, 14853, USA
| | - Liyuan Deng
- Departments of Human Genetics and Pediatrics, McGill University, McGill University Health Center (MUHC), Montreal, H4A 3J1, Canada
| | - Qing Wu
- Departments of Human Genetics and Pediatrics, McGill University, McGill University Health Center (MUHC), Montreal, H4A 3J1, Canada
| | - Zili Zhou
- Departments of Human Genetics and Pediatrics, McGill University, McGill University Health Center (MUHC), Montreal, H4A 3J1, Canada
| | - Zu-Hua Gao
- Department of Pathology, McGill University, Montreal, H4A 3J1, Canada
| | - Pierre Chaurand
- Department of Chemistry, Université de Montréal, Montreal, H3T 1J4, Canada
| | - Marie A Caudill
- Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY, 14853, USA
| | - Rima Rozen
- Departments of Human Genetics and Pediatrics, McGill University, McGill University Health Center (MUHC), Montreal, H4A 3J1, Canada
| |
Collapse
|
24
|
Eshima H, Tamura Y, Kakehi S, Nakamura K, Kurebayashi N, Murayama T, Kakigi R, Sakurai T, Kawamori R, Watada H. Dysfunction of muscle contraction with impaired intracellular Ca 2+ handling in skeletal muscle and the effect of exercise training in male db/db mice. J Appl Physiol (1985) 2018; 126:170-182. [PMID: 30433865 DOI: 10.1152/japplphysiol.00048.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Type 2 diabetes is characterized by reduced contractile force production and increased fatigability of skeletal muscle. While the maintenance of Ca2+ homeostasis during muscle contraction is a requisite for optimal contractile function, the mechanisms underlying muscle contractile dysfunction in type 2 diabetes are unclear. Here, we investigated skeletal muscle contractile force and Ca2+ flux during contraction and pharmacological stimulation in type 2 diabetic model mice ( db/db mice). Furthermore, we investigated the effect of treadmill exercise training on muscle contractile function. In male db/db mice, muscle contractile force and peak Ca2+ levels were both lower during tetanic stimulation of the fast-twitch muscles, while Ca2+ accumulation was higher after stimulation compared with control mice. While 6 wk of exercise training did not improve glucose tolerance, exercise did improve muscle contractile dysfunction, peak Ca2+ levels, and Ca2+ accumulation following stimulation in male db/db mice. These data suggest that dysfunctional Ca2+ flux may contribute to skeletal muscle contractile dysfunction in type 2 diabetes and that exercise training may be a promising therapeutic approach for dysfunctional skeletal muscle contraction. NEW & NOTEWORTHY The purpose of this study was to examine muscle contractile function and Ca2+ regulation as well as the effect of exercise training in skeletal muscle in obese diabetic mice ( db/db). We observed impairment of muscle contractile force and Ca2+ regulation in a male type 2 diabetic animal model. These dysfunctions in muscle were improved by 6 wk of exercise training.
Collapse
Affiliation(s)
- Hiroaki Eshima
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine , Tokyo , Japan.,Sportology Center, Juntendo University Graduate School of Medicine , Tokyo , Japan.,The Japan Society for the Promotion of Science , Tokyo , Japan
| | - Yoshifumi Tamura
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine , Tokyo , Japan.,Sportology Center, Juntendo University Graduate School of Medicine , Tokyo , Japan
| | - Saori Kakehi
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine , Tokyo , Japan.,Sportology Center, Juntendo University Graduate School of Medicine , Tokyo , Japan
| | - Kyoko Nakamura
- Department of Physiology, Juntendo University Graduate School of Medicine , Tokyo , Japan
| | - Nagomi Kurebayashi
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine , Tokyo , Japan
| | - Takashi Murayama
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine , Tokyo , Japan
| | - Ryo Kakigi
- Department of Physiology, Juntendo University Graduate School of Medicine , Tokyo , Japan
| | - Takashi Sakurai
- Department of Cellular and Molecular Pharmacology, Juntendo University Graduate School of Medicine , Tokyo , Japan
| | - Ryuzo Kawamori
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine , Tokyo , Japan.,Sportology Center, Juntendo University Graduate School of Medicine , Tokyo , Japan
| | - Hirotaka Watada
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine , Tokyo , Japan.,Sportology Center, Juntendo University Graduate School of Medicine , Tokyo , Japan.,Center for Therapeutic Innovations in Diabetes, Juntendo University Graduate School of Medicine , Tokyo , Japan.,Center for Molecular Diabetology, Juntendo University Graduate School of Medicine , Tokyo , Japan
| |
Collapse
|
25
|
Rojas JM, Bolze F, Thorup I, Nowak J, Dalsgaard CM, Skydsgaard M, Berthelsen LO, Keane KA, Søeborg H, Sjögren I, Jensen JT, Fels JJ, Offenberg HK, Andersen LW, Dalgaard M. The Effect of Diet-induced Obesity on Toxicological Parameters in the Polygenic Sprague-Dawley Rat Model. Toxicol Pathol 2018; 46:777-798. [DOI: 10.1177/0192623318803557] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | - Florian Bolze
- Toxicology Development Projects, Novo Nordisk A/S, Måløv, Denmark
| | - Inger Thorup
- Toxicopathology, Novo Nordisk A/S, Måløv, Denmark
| | - Jette Nowak
- Toxicopathology, Novo Nordisk A/S, Måløv, Denmark
| | | | | | | | | | | | | | | | | | | | | | - Majken Dalgaard
- Early Regulatory Toxicology, Novo Nordisk A/S, Måløv, Denmark
| |
Collapse
|
26
|
Lubbers ER, Price MV, Mohler PJ. Arrhythmogenic Substrates for Atrial Fibrillation in Obesity. Front Physiol 2018; 9:1482. [PMID: 30405438 PMCID: PMC6204377 DOI: 10.3389/fphys.2018.01482] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 10/01/2018] [Indexed: 12/19/2022] Open
Abstract
Global obesity rates have nearly tripled since 1975. This obesity rate increase is mirrored by increases in atrial fibrillation (AF) that now impacts nearly 10% of Americans over the age of 65. Numerous epidemiologic studies have linked incidence of AF and obesity and other obesity-related diseases, including hypertension and diabetes. Due to the wealth of epidemiologic data linking AF with obesity-related disease, mechanisms of AF pathogenesis in the context of obesity are an area of ongoing investigation. However, progress has been somewhat slowed by the complex phenotype of obesity; separating the effects of obesity from those of related sequelae is problematic. While the initiation of pathogenic pathways leading to AF varies with disease (including increased glycosylation in diabetes, increased renin angiotensin aldosterone system activation in hypertension, atrial ischemia in coronary artery disease, and sleep apnea) the pathogenesis of AF is united by shared mediators of altered conduction in the atria. We suggest focusing on these downstream mediators of AF in obesity is likely to yield more broadly applicable data. In the context of obesity, AF is driven by the interrelated processes of inflammation, atrial remodeling, and oxidative stress. Obesity is characterized by a constant low-grade inflammation that leads to increased expression of pro-inflammatory cytokines. These cytokines contribute to changes in cardiomyocyte excitability. Atrial structural remodeling, including fibrosis, enlargement, and fatty infiltration is a prominent feature of AF and contributes to the altered conduction. Finally, obesity impacts oxidative stress. Within the cardiomyocyte, oxidative stress is increased through both increased production of reactive oxygen species and by downregulation of scavenging enzymes. This increased oxidative stress modulates of cardiomyocyte excitability, increasing susceptibility to AF. Although the initiating insults vary, inflammation, atrial remodeling, and oxidative stress are conserved mechanisms in the pathophysiology of AF in the obese patients. In this review, we highlight mechanisms that have been shown to be relevant in the pathogenesis of AF across obesity-related disease.
Collapse
Affiliation(s)
- Ellen R. Lubbers
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Medical Scientist Training Program, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Morgan V. Price
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Peter J. Mohler
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Physiology & Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
27
|
He C, Hu X, Weston TA, Jung RS, Heizer P, Tu Y, Ellison R, Matsumoto K, Gerhardt H, Tontonoz P, Fong LG, Young SG, Jiang H. NanoSIMS imaging reveals unexpected heterogeneity in nutrient uptake by brown adipocytes. Biochem Biophys Res Commun 2018; 504:899-902. [PMID: 30224066 DOI: 10.1016/j.bbrc.2018.09.051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 09/08/2018] [Indexed: 11/16/2022]
Abstract
Heterogeneity in the metabolic properties of adipocytes in white adipose tissue has been well documented. We sought to investigate metabolic heterogeneity in adipocytes of brown adipose tissue (BAT), focusing on heterogeneity in nutrient uptake. To explore the possibility of metabolic heterogeneity in brown adipocytes, we used nanoscale secondary ion mass spectrometry (NanoSIMS) to quantify uptake of lipids in adipocytes interscapular BAT and perivascular adipose tissue (PVAT) after an intravenous injection of triglyceride-rich lipoproteins (TRLs) containing [2H]triglycerides (2H-TRLs). The uptake of deuterated lipids into brown adipocytes was quantified by NanoSIMS. We also examined 13C enrichment in brown adipocytes after administering [13C]glucose or 13C-labeled mixed fatty acids by gastric gavage. The uptake of 2H-TRLs-derived lipids into brown adipocytes was heterogeneous, with 2H enrichment in adjacent adipocytes varying by more than fourfold. We also observed substantial heterogeneity in 13C enrichment in adjacent brown adipocytes after administering [13C]glucose or [13C]fatty acids by gastric gavage. The uptake of nutrients by adjacent brown adipocytes within a single depot is variable, suggesting that there is heterogeneity in the metabolic properties of brown adipocytes.
Collapse
Affiliation(s)
- Cuiwen He
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Xuchen Hu
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Thomas A Weston
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Rachel S Jung
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Patrick Heizer
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Yiping Tu
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Rochelle Ellison
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Ken Matsumoto
- VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium
| | - Holger Gerhardt
- VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium; Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Peter Tontonoz
- Departments of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Loren G Fong
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA.
| | - Stephen G Young
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA; Departments of Human Genetics, University of California, Los Angeles, CA, 90095, USA.
| | - Haibo Jiang
- Departments of Medicine, University of California, Los Angeles, CA, 90095, USA; School of Molecular Sciences and Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Perth, 6009, Australia.
| |
Collapse
|
28
|
Hesse D, Trost J, Schäfer N, Schwerbel K, Hoeflich A, Schürmann A, Brockmann GA. Effect of adipocyte-derived IGF-I on adipose tissue mass and glucose metabolism in the Berlin Fat Mouse. Growth Factors 2018; 36:78-88. [PMID: 30196772 DOI: 10.1080/08977194.2018.1497621] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Besides liver, IGF-I is expressed in adipose tissue. However, the effects of this local IGF-I on adipose tissue and metabolism are unclear. We generated adipocyte-specific knock-out mice on the background of the Berlin Fat Mouse Inbred (BFMI) line to evaluate the contribution of adipocyte-IGF-I on glucose metabolism and adipose tissue development. BFMI mice are obese, non-diabetic with elevated plasma insulin and IGF-I concentration. The knock-out in adipocytes led to a total white adipose tissue expression of 50-60% due to unaltered Igf-1 expression in stromavascular cells. The lack of IGF-I from adipocytes did not alter plasma IGF-I concentration. BFMIChr3-Igf-I-KOQ-AT mice had reduced adipose tissue mass in most depots. During oral glucose tolerance tests, BFMIChr3-Igf-I-KOQ-AT mice showed an impaired glucose clearance (p = .03). Interestingly, insulin action was enhanced during insulin tolerance tests (p = .05). In conclusion, adipocyte-specific IGF-I ablation in obese BFMI mice results in reduced adipose tissue mass and thereby alters glucose metabolism.
Collapse
Affiliation(s)
- Deike Hesse
- a Department of Crop and Animal Sciences, Humboldt-Universität zu Berlin , Berlin , Germany
| | - Jan Trost
- a Department of Crop and Animal Sciences, Humboldt-Universität zu Berlin , Berlin , Germany
| | - Nadine Schäfer
- a Department of Crop and Animal Sciences, Humboldt-Universität zu Berlin , Berlin , Germany
| | - Kristin Schwerbel
- b German Institute of Human Nutrition , Nuthetal , Germany
- c German Center for Diabetes Research , München-Neuherberg , Germany
| | - Andreas Hoeflich
- d Leibniz Institute for Farm Animal Biology , Dummerstorf , Germany
| | - Annette Schürmann
- b German Institute of Human Nutrition , Nuthetal , Germany
- c German Center for Diabetes Research , München-Neuherberg , Germany
| | - Gudrun A Brockmann
- a Department of Crop and Animal Sciences, Humboldt-Universität zu Berlin , Berlin , Germany
| |
Collapse
|
29
|
Russell PK, Mangiafico S, Fam BC, Clarke MV, Marin ES, Andrikopoulos S, Wiren KM, Zajac JD, Davey RA. The androgen receptor in bone marrow progenitor cells negatively regulates fat mass. J Endocrinol 2018; 237:15-27. [PMID: 29386237 DOI: 10.1530/joe-17-0656] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 01/29/2018] [Indexed: 12/14/2022]
Abstract
It is well established that testosterone negatively regulates fat mass in humans and mice; however, the mechanism by which testosterone exerts these effects is poorly understood. We and others have shown that deletion of the androgen receptor (AR) in male mice results in a phenotype that mimics the three key clinical aspects of hypogonadism in human males; increased fat mass and decreased bone and muscle mass. We now show that replacement of the Ar gene specifically in mesenchymal progenitor cells (PCs) residing in the bone marrow of Global-ARKO mice, in the absence of the AR in all other tissues (PC-AR Gene Replacements), completely attenuates their increased fat accumulation. Inguinal subcutaneous white adipose tissue and intra-abdominal retroperitoneal visceral adipose tissue depots in PC-AR Gene Replacement mice were 50-80% lower than wild-type (WT) and 75-90% lower than Global-ARKO controls at 12 weeks of age. The marked decrease in subcutaneous and visceral fat mass in PC-AR Gene Replacements was associated with an increase in the number of small adipocytes and a healthier metabolic profile compared to WT controls, characterised by normal serum leptin and elevated serum adiponectin levels. Euglycaemic/hyperinsulinaemic clamp studies reveal that the PC-AR Gene Replacement mice have improved whole-body insulin sensitivity with higher glucose infusion rates compared to WT mice and increased glucose uptake into subcutaneous and intra-abdominal fat. In conclusion, these data provide the first evidence for an action of androgens via the AR in mesenchymal bone marrow PCs to negatively regulate fat mass and improve metabolic function.
Collapse
Affiliation(s)
- Patricia K Russell
- Department of MedicineAustin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Salvatore Mangiafico
- Department of MedicineAustin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Barbara C Fam
- Department of MedicineAustin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Michele V Clarke
- Department of MedicineAustin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Evelyn S Marin
- Department of MedicineAustin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Sofianos Andrikopoulos
- Department of MedicineAustin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Kristine M Wiren
- Research ServiceVeterans Affairs Medical Center, Portland, Oregon, USA
- Departments of Medicine and Behavioral NeuroscienceOregon Health & Science University, Portland, Oregon, USA
| | - Jeffrey D Zajac
- Department of MedicineAustin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Rachel A Davey
- Department of MedicineAustin Health, University of Melbourne, Heidelberg, Victoria, Australia
| |
Collapse
|
30
|
Schoettl T, Fischer IP, Ussar S. Heterogeneity of adipose tissue in development and metabolic function. ACTA ACUST UNITED AC 2018. [PMID: 29514879 DOI: 10.1242/jeb.162958] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adipose tissue is a central metabolic organ. Unlike other organs, adipose tissue is compartmentalized into individual depots and distributed throughout the body. These different adipose depots show major functional differences and risk associations for developing metabolic syndrome. Recent advances in lineage tracing demonstrate that individual adipose depots are composed of adipocytes that are derived from distinct precursor populations, giving rise to different populations of energy-storing white adipocytes. Moreover, distinct lineages of energy-dissipating brown and beige adipocytes exist in discrete depots or within white adipose tissue depots. In this Review, we discuss developmental and functional heterogeneity, as well as sexual dimorphism, between and within individual adipose tissue depots. We highlight current data relating to the differences between subcutaneous and visceral white adipose tissue in the development of metabolic dysfunction, with special emphasis on adipose tissue expansion and remodeling of the extracellular matrix. Moreover, we provide a detailed overview of adipose tissue development as well as the consensus and controversies relating to adult adipocyte precursor populations.
Collapse
Affiliation(s)
- Theresa Schoettl
- JRG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, 85748 Garching, Germany.,German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Ingrid P Fischer
- JRG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, 85748 Garching, Germany.,German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany.,Division of Metabolic Diseases, Department of Medicine, Technische Universität München, 80333 Munich, Germany
| | - Siegfried Ussar
- JRG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, 85748 Garching, Germany .,German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| |
Collapse
|
31
|
Depot-specific inflammation with decreased expression of ATM2 in white adipose tissues induced by high-margarine/lard intake. PLoS One 2017; 12:e0188007. [PMID: 29141038 PMCID: PMC5687764 DOI: 10.1371/journal.pone.0188007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 10/30/2017] [Indexed: 01/21/2023] Open
Abstract
A high-fat diet has been recognized as an important risk factor of obesity, with variable impacts of different fatty acid compositions on the physiological process. To understand the effects of a high-margarine/lard diet, which is a major source of trans fatty acids (TFAs)/ saturated fatty acids (SFAs), elaidic acid as a biomarker of margarine intake was used to screen affected adipokines on mature human adipocytes in vitro. Weaned male Wistar rats were fed a high-fat diet enriched with margarine/lard to generate obesity-prone (OP) and obesity-resistant (OR) models, which were then used to explore the inflammatory responses of depot-specific white adipose tissue. Adiposity, glucose and lipid metabolism parameters and macrophage cell markers were also compared in vivo. In the subcutaneous depot, a high-margarine diet induced elevated IL-6, MCP-1 and XCL1 expression levels in both M-OP and M-OR groups. High-lard diet-fed rats displayed higher protein expression levels of MCP-1 and XCL1 compared with the control group. In the epididymal depot, significantly elevated IL-6 production was observed in M-OP rats, and high-lard diet-fed rats displayed elevated IL-6 and decreased XCL1 expression. In the retroperitoneal depot, a high-margarine diet caused higher IL-6 and MCP-1 expression levels, a high-lard diet caused elevated IL-6 expression in L-OP/L-OR rats, and elevated XCL1 expression was observed only in L-OP rats. In general, CD206 mRNA levels were notably down-regulated by high-fat diet feeding in the above-mentioned depots. CD11c mRNA levels were slightly upregulated in the subcutaneous depot of OP rats fed a high-margarine/lard diet. In the epidydimal depot, higher expression levels of F4/80 and CD206 mRNA were observed only in high-margarine diet-fed OP rats. These results suggest that depot-specific inflammation with decreased expression of adipose tissue anti-inflammatory M2-type (ATM2) macrophages could be induced by high-margarine/lard intake.
Collapse
|
32
|
Berryman DE, List EO. Growth Hormone's Effect on Adipose Tissue: Quality versus Quantity. Int J Mol Sci 2017; 18:ijms18081621. [PMID: 28933734 PMCID: PMC5578013 DOI: 10.3390/ijms18081621] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 07/10/2017] [Accepted: 07/17/2017] [Indexed: 02/07/2023] Open
Abstract
Obesity is an excessive accumulation or expansion of adipose tissue (AT) due to an increase in either the size and/or number of its characteristic cell type, the adipocyte. As one of the most significant public health problems of our time, obesity and its associated metabolic complications have demanded that attention be given to finding effective therapeutic options aimed at reducing adiposity or the metabolic dysfunction associated with its accumulation. Growth hormone (GH) has therapeutic potential due to its potent lipolytic effect and resultant ability to reduce AT mass while preserving lean body mass. However, AT and its resident adipocytes are significantly more dynamic and elaborate than once thought and require one not to use the reduction in absolute mass as a readout of efficacy alone. Paradoxically, therapies that reduce GH action may ultimately prove to be healthier, in part because GH also possesses potent anti-insulin activities along with concerns that GH may promote the growth of certain cancers. This review will briefly summarize some of the newer complexities of AT relevant to GH action and describe the current understanding of how GH influences this tissue using data from both humans and mice. We will conclude by considering the therapeutic use of GH or GH antagonists in obesity, as well as important gaps in knowledge regarding GH and AT.
Collapse
Affiliation(s)
- Darlene E Berryman
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, OH 45701, USA.
- Edison Biotechnology Institute, 218 Konneker Research Labs, Ohio University, Athens, OH 45701, USA.
| | - Edward O List
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, OH 45701, USA.
- Edison Biotechnology Institute, 218 Konneker Research Labs, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
33
|
Duran-Ortiz S, Brittain AL, Kopchick JJ. The impact of growth hormone on proteomic profiles: a review of mouse and adult human studies. Clin Proteomics 2017; 14:24. [PMID: 28670222 PMCID: PMC5492507 DOI: 10.1186/s12014-017-9160-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 06/20/2017] [Indexed: 12/17/2022] Open
Abstract
Growth hormone (GH) is a protein that is known to stimulate postnatal growth, counter regulate insulin's action and induce expression of insulin-like growth factor-1. GH exerts anabolic or catabolic effects depending upon on the targeted tissue. For instance, GH increases skeletal muscle and decreases adipose tissue mass. Our laboratory has spent the past two decades studying these effects, including the effects of GH excess and depletion, on the proteome of several mouse and human tissues. This review first discusses proteomic techniques that are commonly used for these types of studies. We then examine the proteomic differences found in mice with excess circulating GH (bGH mice) or mice with disruption of the GH receptor gene (GHR-/-). We also describe the effects of increased and decreased GH action on the proteome of adult patients with either acromegaly, GH deficiency or patients after short-term GH treatment. Finally, we explain how these proteomic studies resulted in the discovery of potential biomarkers for GH action, particularly those related with the effects of GH on aging, glucose metabolism and body composition.
Collapse
Affiliation(s)
- Silvana Duran-Ortiz
- Edison Biotechnology Institute, Ohio University, Athens, OH USA.,Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH USA.,Molecular and Cellular Biology Program, Ohio University, Athens, OH USA
| | - Alison L Brittain
- Edison Biotechnology Institute, Ohio University, Athens, OH USA.,Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, OH USA.,Molecular and Cellular Biology Program, Ohio University, Athens, OH USA.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701 USA
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH USA.,Molecular and Cellular Biology Program, Ohio University, Athens, OH USA.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701 USA
| |
Collapse
|
34
|
Troike KM, Henry BE, Jensen EA, Young JA, List EO, Kopchick JJ, Berryman DE. Impact of Growth Hormone on Regulation of Adipose Tissue. Compr Physiol 2017. [PMID: 28640444 DOI: 10.1002/cphy.c160027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Increasing prevalence of obesity and obesity-related conditions worldwide has necessitated a more thorough understanding of adipose tissue (AT) and expanded the scope of research in this field. AT is now understood to be far more complex and dynamic than previously thought, which has also fueled research to reevaluate how hormones, such as growth hormone (GH), alter the tissue. In this review, we will introduce properties of AT important for understanding how GH alters the tissue, such as anatomical location of depots and adipokine output. We will provide an overview of GH structure and function and define several human conditions and cognate mouse lines with extremes in GH action that have helped shape our understanding of GH and AT. A detailed discussion of the GH/AT relationship will be included that addresses adipokine production, immune cell populations, lipid metabolism, senescence, differentiation, and fibrosis, as well as brown AT and beiging of white AT. A brief overview of how GH levels are altered in an obese state, and the efficacy of GH as a therapeutic option to manage obesity will be given. As we will reveal, the effects of GH on AT are numerous, dynamic and depot-dependent. © 2017 American Physiological Society. Compr Physiol 7:819-840, 2017.
Collapse
Affiliation(s)
- Katie M Troike
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,School of Applied Health Sciences and Wellness, College of Health Sciences and Professions, Ohio University, Athens, Ohio, USA
| | - Brooke E Henry
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,School of Applied Health Sciences and Wellness, College of Health Sciences and Professions, Ohio University, Athens, Ohio, USA
| | - Elizabeth A Jensen
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - Jonathan A Young
- Department of Biological Sciences, College of Arts and Sciences, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - Edward O List
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - John J Kopchick
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA.,Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, Ohio, USA
| | - Darlene E Berryman
- The Diabetes Institute at Ohio University, 108 Konneker Research Labs, Ohio University, Athens, Ohio, USA.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| |
Collapse
|
35
|
Boumelhem BB, Assinder SJ, Bell-Anderson KS, Fraser ST. Flow cytometric single cell analysis reveals heterogeneity between adipose depots. Adipocyte 2017; 6:112-123. [PMID: 28453382 DOI: 10.1080/21623945.2017.1319536] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Understanding adipose tissue heterogeneity is hindered by the paucity of methods to analyze mature adipocytes at the single cell level. Here, we report a system for analyzing live adipocytes from different adipose depots in the adult mouse. Single cell suspensions of buoyant adipocytes were separated from the stromal vascular fraction and analyzed by flow cytometry. Compared to other lipophilic dyes, Nile Red uptake effectively distinguished adipocyte populations. Nile Red fluorescence increased with adipocyte size and granularity and could be combined with MitoTracker® Deep Red or fluorescent antibody labeling to further dissect adipose populations. Epicardial adipocytes exhibited the least mitochondrial membrane depolarization and highest fatty-acid translocase CD36 surface expression. In contrast, brown adipocytes showed low surface CD36 expression. Pregnancy resulted in reduced mitochondrial membrane depolarisation and increased CD36 surface expression in brown and epicardial adipocyte populations respectively. Our protocol revealed unreported heterogeneity between adipose depots and highlights the utility of flow cytometry for screening adipocytes at the single cell level.
Collapse
Affiliation(s)
- Badwi B. Boumelhem
- Discipline of Physiology, University of Sydney, Sydney, Australia
- Bosch Institute, University of Sydney, Sydney, Australia
| | - Stephen J. Assinder
- Discipline of Physiology, University of Sydney, Sydney, Australia
- Bosch Institute, University of Sydney, Sydney, Australia
| | - Kim S. Bell-Anderson
- Bosch Institute, University of Sydney, Sydney, Australia
- Discipline of Anatomy and Histology, University of Sydney, Sydney, Australia
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, Australia
| | - Stuart T. Fraser
- Discipline of Physiology, University of Sydney, Sydney, Australia
- Bosch Institute, University of Sydney, Sydney, Australia
- Discipline of Anatomy and Histology, University of Sydney, Sydney, Australia
| |
Collapse
|
36
|
Raciti GA, Spinelli R, Desiderio A, Longo M, Parrillo L, Nigro C, D'Esposito V, Mirra P, Fiory F, Pilone V, Forestieri P, Formisano P, Pastan I, Miele C, Beguinot F. Specific CpG hyper-methylation leads to Ankrd26 gene down-regulation in white adipose tissue of a mouse model of diet-induced obesity. Sci Rep 2017; 7:43526. [PMID: 28266632 PMCID: PMC5339897 DOI: 10.1038/srep43526] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 01/27/2017] [Indexed: 12/16/2022] Open
Abstract
Epigenetic modifications alter transcriptional activity and contribute to the effects of environment on the individual risk of obesity and Type 2 Diabetes (T2D). Here, we have estimated the in vivo effect of a fat-enriched diet (HFD) on the expression and the epigenetic regulation of the Ankyrin repeat domain 26 (Ankrd26) gene, which is associated with the onset of these disorders. In visceral adipose tissue (VAT), HFD exposure determined a specific hyper-methylation of Ankrd26 promoter at the −436 and −431 bp CpG sites (CpGs) and impaired its expression. Methylation of these 2 CpGs impaired binding of the histone acetyltransferase/transcriptional coactivator p300 to this same region, causing hypo-acetylation of histone H4 at the Ankrd26 promoter and loss of binding of RNA Pol II at the Ankrd26 Transcription Start Site (TSS). In addition, HFD increased binding of DNA methyl-transferases (DNMTs) 3a and 3b and methyl-CpG-binding domain protein 2 (MBD2) to the Ankrd26 promoter. More importantly, Ankrd26 down-regulation enhanced secretion of pro-inflammatory mediators by 3T3-L1 adipocytes as well as in human sera. Thus, in mice, the exposure to HFD induces epigenetic silencing of the Ankrd26 gene, which contributes to the adipose tissue inflammatory secretion profile induced by high-fat regimens.
Collapse
Affiliation(s)
- Gregory A Raciti
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Rosa Spinelli
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Antonella Desiderio
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Michele Longo
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Luca Parrillo
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Cecilia Nigro
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Vittoria D'Esposito
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Paola Mirra
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Francesca Fiory
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Vincenzo Pilone
- Bariatric and Metabolic Surgery Unit, University of Salerno, Salerno, 84084, Italy
| | - Pietro Forestieri
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, 80131, Italy
| | - Pietro Formisano
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Ira Pastan
- Laboratory of Molecular Biology (LMB), National Cancer Institute (NCI), National Institute of Health (NIH), Bethesda, MD 20892, USA
| | - Claudia Miele
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| | - Francesco Beguinot
- URT of the Institute of Experimental Endocrinology and Oncology "G. Salvatore", National Council of Research, Naples, 80131, Italy.,Department of Translational Medical Sciences, University of Naples "Federico II", Naples, 80131, Italy
| |
Collapse
|
37
|
Danneskiold-Samsøe NB, Andersen D, Radulescu ID, Normann-Hansen A, Brejnrod A, Kragh M, Madsen T, Nielsen C, Josefsen K, Fretté X, Fjaere E, Madsen L, Hellgren LI, Brix S, Kristiansen K. A safflower oil based high-fat/high-sucrose diet modulates the gut microbiota and liver phospholipid profiles associated with early glucose intolerance in the absence of tissue inflammation. Mol Nutr Food Res 2017; 61. [DOI: 10.1002/mnfr.201600528] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 11/15/2016] [Accepted: 12/12/2016] [Indexed: 01/15/2023]
Affiliation(s)
| | - Daniel Andersen
- Department of Biotechnology and Biomedicine; Technical University of Denmark; Lyngby Denmark
| | - Ilinca Daria Radulescu
- Rigshospitalet Department 3733; Copenhagen Biocenter; The Bartholin Institute; Copenhagen Denmark
| | - Ann Normann-Hansen
- Laboratory of Genomics and Molecular Biomedicine; Department of Biology; University of Copenhagen; Copenhagen Denmark
| | - Asker Brejnrod
- Laboratory of Genomics and Molecular Biomedicine; Department of Biology; University of Copenhagen; Copenhagen Denmark
| | - Marie Kragh
- Department of Biotechnology and Biomedicine; Technical University of Denmark; Lyngby Denmark
| | - Tobias Madsen
- Department of Biotechnology and Biomedicine; Technical University of Denmark; Lyngby Denmark
| | - Christian Nielsen
- Department of Biotechnology and Biomedicine; Technical University of Denmark; Lyngby Denmark
| | - Knud Josefsen
- Rigshospitalet Department 3733; Copenhagen Biocenter; The Bartholin Institute; Copenhagen Denmark
| | - Xavier Fretté
- Department of Chemical Engineering; Biotechnology and Environmental Technology; University of Southern Denmark; Odense M Denmark
| | - Even Fjaere
- National Institute of Nutrition and Seafood Research; Bergen Norway
| | - Lise Madsen
- Laboratory of Genomics and Molecular Biomedicine; Department of Biology; University of Copenhagen; Copenhagen Denmark
- National Institute of Nutrition and Seafood Research; Bergen Norway
| | - Lars I. Hellgren
- Department of Biotechnology and Biomedicine; Technical University of Denmark; Lyngby Denmark
| | - Susanne Brix
- Department of Biotechnology and Biomedicine; Technical University of Denmark; Lyngby Denmark
| | - Karsten Kristiansen
- Laboratory of Genomics and Molecular Biomedicine; Department of Biology; University of Copenhagen; Copenhagen Denmark
- BGI-Shenzhen; Shenzhen China
| |
Collapse
|
38
|
Zhu Q, Ghoshal S, Tyagi R, Chakraborty A. Global IP6K1 deletion enhances temperature modulated energy expenditure which reduces carbohydrate and fat induced weight gain. Mol Metab 2016; 6:73-85. [PMID: 28123939 PMCID: PMC5220553 DOI: 10.1016/j.molmet.2016.11.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 11/15/2016] [Accepted: 11/23/2016] [Indexed: 01/27/2023] Open
Abstract
OBJECTIVE IP6 kinases (IP6Ks) regulate cell metabolism and survival. Mice with global (IP6K1-KO) or adipocyte-specific (AdKO) deletion of IP6K1 are protected from diet induced obesity (DIO) at ambient (23 °C) temperature. AdKO mice are lean primarily due to increased AMPK mediated thermogenic energy expenditure (EE). Thus, at thermoneutral (30 °C) temperature, high fat diet (HFD)-fed AdKO mice expend energy and gain body weight, similar to control mice. IP6K1 is ubiquitously expressed; thus, it is critical to determine to what extent the lean phenotype of global IP6K1-KO mice depends on environmental temperature. Furthermore, it is not known whether IP6K1 regulates AMPK mediated EE in cells, which do not express UCP1. METHODS Q-NMR, GTT, food intake, EE, QRT-PCR, histology, mitochondrial oxygen consumption rate (OCR), fatty acid metabolism assays, and immunoblot studies were conducted in IP6K1-KO and WT mice or cells. RESULTS Global IP6K1 deletion mediated enhancement in EE is impaired albeit not abolished at 30 °C. As a result, IP6K1-KO mice are protected from DIO, insulin resistance, and fatty liver even at 30 °C. Like AdKO, IP6K1-KO mice display enhanced adipose tissue browning. However, unlike AdKO mice, thermoneutrality only partly abolishes browning in IP6K1-KO mice. Cold (5 °C) exposure enhances carbohydrate expenditure, whereas 23 °C and 30 °C promote fat oxidation in HFD-KO mice. Furthermore, IP6K1 deletion diminishes cellular fat accumulation via activation of the AMPK signaling pathway. CONCLUSIONS Global deletion of IP6K1 ameliorates obesity and insulin resistance irrespective of the environmental temperature conditions, which strengthens its validity as an anti-obesity target.
Collapse
Affiliation(s)
- Qingzhang Zhu
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL, 33458, USA
| | - Sarbani Ghoshal
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL, 33458, USA
| | - Richa Tyagi
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Anutosh Chakraborty
- Department of Metabolism and Aging, The Scripps Research Institute, Jupiter, FL, 33458, USA.
| |
Collapse
|
39
|
Yamada LTP, de Oliveira MC, Batista NV, Fonseca RC, Sousa Pereira RV, Perez DA, Teixeira MM, Cara DC, Ferreira AVM. Immunologic and metabolic effects of high-refined carbohydrate-containing diet in food allergic mice. Nutrition 2016; 32:273-80. [DOI: 10.1016/j.nut.2015.08.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 07/20/2015] [Accepted: 08/14/2015] [Indexed: 01/21/2023]
|
40
|
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: 2.1] [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
|
41
|
Sierra Rojas JX, García-San Frutos M, Horrillo D, Lauzurica N, Oliveros E, Carrascosa JM, Fernández-Agulló T, Ros M. Differential Development of Inflammation and Insulin Resistance in Different Adipose Tissue Depots Along Aging in Wistar Rats: Effects of Caloric Restriction. J Gerontol A Biol Sci Med Sci 2015; 71:310-22. [DOI: 10.1093/gerona/glv117] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 02/14/2015] [Indexed: 01/06/2023] Open
|
42
|
Krueger KC, Feldman BJ. Adipose circadian clocks: coordination of metabolic rhythms by clock genes, steroid hormones, and PPARs. Horm Mol Biol Clin Investig 2015; 14:15-24. [PMID: 25436716 DOI: 10.1515/hmbci-2013-0011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 04/30/2013] [Indexed: 12/23/2022]
Abstract
A central clock consisting of interconnected positive and negative feedback gene loops operates in the brain, tying rhythmic activity to the 24-h day. The central clock entrains similar feedback loops present in most peripheral tissues to coordinate metabolic gene expression among organs and with feeding activity for more efficient utilization of resources. Recent studies are beginning to elucidate the intricate feedback mechanisms among central and peripheral clocks and their roles in activity and metabolic homeostasis. Adipose tissue serves as a major energy storage organ and releases paracrine and endocrine hormones to signal energy status to other organs. Within the adipose tissue, the transcriptional feedback regulation between clock genes and nuclear hormone receptors, together with direct protein associations among these molecules, ensures the expression of metabolic genes at the appropriate time. This review will summarize the important components and mechanisms of adipose clock entrainment, particularly highlighting instructive studies carried out in mice. This research not only illustrates the intricate connections between clocks and metabolism but also provides potential mechanisms to correct abnormalities induced by disrupted sleep or poor diet.
Collapse
|
43
|
van Beek L, van Klinken JB, Pronk ACM, van Dam AD, Dirven E, Rensen PCN, Koning F, Willems van Dijk K, van Harmelen V. The limited storage capacity of gonadal adipose tissue directs the development of metabolic disorders in male C57Bl/6J mice. Diabetologia 2015; 58:1601-9. [PMID: 25962520 PMCID: PMC4473015 DOI: 10.1007/s00125-015-3594-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 03/31/2015] [Indexed: 12/26/2022]
Abstract
AIMS/HYPOTHESIS White adipose tissue (WAT) consists of various depots with different adipocyte functionality and immune cell composition. Knowledge of WAT-depot-specific differences in expandability and immune cell influx during the development of obesity is limited, therefore we aimed to characterise different WAT depots during the development of obesity in mice. METHODS Gonadal WAT (gWAT), subcutaneous WAT (sWAT) and mesenteric WAT (mWAT) were isolated from male C57Bl/6J mice with different body weights (approximately 25-60 g) and analysed. Linear and non-linear regression models were used to describe the extent of WAT depot expandability and immune cell composition as a function of body weight. RESULTS Whereas mouse sWAT and mWAT continued to expand with body weight, gWAT expanded mainly during the initial phase of body weight gain. The expansion diminished after the mice reached a body weight of around 40 g. From this point on, gWAT crown-like structure formation, liver steatosis and insulin resistance occurred. Mouse WAT depots showed major differences in immune cell composition: gWAT consisted mainly of macrophages, whereas sWAT and mWAT primarily contained lymphocytes. CONCLUSIONS/INTERPRETATION Marked inter-depot differences exist in WAT immune cell composition and expandability. The limited storage capacity of gWAT seems to direct the development of metabolic disorders in male C57Bl/6J mice.
Collapse
Affiliation(s)
- Lianne van Beek
- />Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, PO Box 9600, 2300 RC Leiden, the Netherlands
- />Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Jan B. van Klinken
- />Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, PO Box 9600, 2300 RC Leiden, the Netherlands
- />Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Amanda C. M. Pronk
- />Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, PO Box 9600, 2300 RC Leiden, the Netherlands
- />Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Andrea D. van Dam
- />Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
- />Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| | - Eline Dirven
- />Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, PO Box 9600, 2300 RC Leiden, the Netherlands
- />Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Patrick C. N. Rensen
- />Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
- />Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| | - Frits Koning
- />Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Ko Willems van Dijk
- />Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, PO Box 9600, 2300 RC Leiden, the Netherlands
- />Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
- />Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| | - Vanessa van Harmelen
- />Department of Human Genetics, Leiden University Medical Center, Einthovenweg 20, PO Box 9600, 2300 RC Leiden, the Netherlands
- />Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
44
|
During MJ, Liu X, Huang W, Magee D, Slater A, McMurphy T, Wang C, Cao L. Adipose VEGF Links the White-to-Brown Fat Switch With Environmental, Genetic, and Pharmacological Stimuli in Male Mice. Endocrinology 2015; 156:2059-73. [PMID: 25763639 PMCID: PMC4430610 DOI: 10.1210/en.2014-1905] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Living in an enriched environment (EE) decreases adiposity, increases energy expenditure, causes resistance to diet induced obesity, and induces brown-like (beige) cells in white fat via activating a hypothalamic-adipocyte axis. Here we report that EE stimulated vascular endothelial growth factor (VEGF) expression in a fat depot-specific manner prior to the emergence of beige cells. The VEGF up-regulation was independent of hypoxia but required intact sympathetic tone to the adipose tissue. Targeted adipose overexpression of VEGF reproduced the browning effect of EE. Adipose-specific VEGF knockout or pharmacological VEGF blockade with antibodies abolished the induction of beige cell by EE. Hypothalamic brain-derived neurotrophic factor stimulated by EE regulated the adipose VEGF expression, and VEGF signaling was essential to the hypothalamic brain-derived neurotrophic factor-induced white adipose tissue browning. Furthermore, VEGF signaling was essential to the beige cells induction by exercise, a β3-adrenergic agonist, and a peroxisome proliferator-activated receptor-γ ligand, suggesting a common downstream pathway integrating diverse upstream mechanisms. Exploiting this pathway may offer potential therapeutic interventions to obesity and metabolic diseases.
Collapse
Affiliation(s)
- Matthew J During
- Department of Molecular Virology, Immunology, and Medical Genetics and The Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Obesity is associated with altered gut microbiota composition and impaired gut barrier function. These changes, together with interrelated mesenteric adipose tissue inflammation, result in increased release of pro-inflammatory cytokines, bacteria-derived factors, and lipids into the portal circulation, promoting the development of (hepatic) insulin resistance. Herein, the potential impact of obesity-related changes in gut and visceral adipose tissue biology on the development of insulin resistance and Type 2 diabetes is reviewed.
Collapse
Affiliation(s)
- Daniel Konrad
- Department of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland; Children's Research Center, University Children's Hospital, Zurich, Switzerland; and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Stephan Wueest
- Department of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland; Children's Research Center, University Children's Hospital, Zurich, Switzerland; and
| |
Collapse
|
46
|
Benencia F, Harshman S, Duran-Ortiz S, Lubbers ER, List EO, Householder L, Al-Naeeli M, Liang X, Welch L, Kopchick JJ, Berryman DE. Male bovine GH transgenic mice have decreased adiposity with an adipose depot-specific increase in immune cell populations. Endocrinology 2015; 156:1794-803. [PMID: 25521584 PMCID: PMC4398765 DOI: 10.1210/en.2014-1794] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
White adipose tissue (WAT) is composed of mature adipocytes and a stromal vascular fraction (SVF), which contains a variety of cells, including immune cells that vary among the different WAT depots. Growth hormone (GH) impacts immune function and adiposity in an adipose depot-specific manner. However, its effects on WAT immune cell populations remain unstudied. Bovine GH transgenic (bGH) mice are commonly used to study the in vivo effects of GH. These giant mice have an excess of GH action, impaired glucose metabolism, decreased adiposity, increased lean mass, and a shortened lifespan. Therefore, the purpose of this study was to characterize the WAT depot-specific differences in immune cell populations in the presence of excess GH in vivo. Three WAT depots were assessed: inguinal (sc), epididymal (EPI), and mesenteric (MES). Subcutaneous and MES bGH WAT depots showed a significantly higher number of total SVF cells, yet only MES bGH WAT had higher leukocyte counts compared with control samples. By means of flow cytometry analysis of the SVF, we detected greater macrophage and regulatory T-cell infiltration in sc and MES bGH WAT depots compared with controls. However, no differences were observed in the EPI WAT depot. RNA-sequencing confirmed significant alterations in pathways related to T-cell infiltration and activation in the sc depot with fewer significant changes in the EPI bGH WAT depot. These findings collectively point to a previously unrecognized role for GH in influencing the distribution of WAT immune cell populations in a depot-specific manner.
Collapse
Affiliation(s)
- Fabian Benencia
- Department of Biomedical Sciences (F.B., J.J.K., D.E.B.), Heritage College of Osteopathic Medicine; Russ College of Engineering and Technology (F.B.); Diabetes Institute (F.B., E.O.L., M.A.-N., J.J.K., D.E.B.); Edison Biotechnology Institute (S.H., S.D.-O., E.R.L., E.O.L., L.H., J.J.K., D.E.B.); School of Applied Health Sciences and Wellness (S.H., S.D.-O., D.E.B.), College of Health Sciences and Professions; Department of Biological Sciences (M.A.-N.), Ohio University Zanesville; School of Electrical Engineering and Computer Science (X.L., L.W.); and Biomedical Engineering Program (L.W.), Ohio University, Athens, Ohio 45701
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
van der Stelt I, Hoek-van den Hil EF, Swarts HJ, Vervoort JJ, Hoving L, Skaltsounis L, Lemonakis N, Andreadou I, van Schothorst EM, Keijer J. Nutraceutical oleuropein supplementation prevents high fat diet-induced adiposity in mice. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.02.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
48
|
Schöttl T, Kappler L, Braun K, Fromme T, Klingenspor M. Limited mitochondrial capacity of visceral versus subcutaneous white adipocytes in male C57BL/6N mice. Endocrinology 2015; 156:923-33. [PMID: 25549046 DOI: 10.1210/en.2014-1689] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Accumulation of visceral fat is associated with metabolic risk whereas excessive amounts of peripheral fat are considered less problematic. At the same time, altered white adipocyte mitochondrial bioenergetics has been implicated in the pathogenesis of insulin resistance and type 2 diabetes. We therefore investigated whether the metabolic risk of visceral vs peripheral fat coincides with a difference in mitochondrial capacity of white adipocytes. We assessed bioenergetic parameters of subcutaneous inguinal and visceral epididymal white adipocytes from male C57BL/6N mice employing a comprehensive respirometry setup of intact and permeabilized adipocytes as well as isolated mitochondria. Inguinal adipocytes clearly featured a higher respiratory capacity attributable to increased mitochondrial respiratory chain content compared with epididymal adipocytes. The lower capacity of mitochondria from epididymal adipocytes was accompanied by an increased generation of reactive oxygen species per oxygen consumed. Feeding a high-fat diet (HFD) for 1 week reduced white adipocyte mitochondrial capacity, with stronger effects in epididymal when compared with inguinal adipocytes. This was accompanied by impaired body glucose homeostasis. Therefore, the limited bioenergetic performance combined with the proportionally higher generation of reactive oxygen species of visceral adipocytes could be seen as a candidate mechanism mediating the elevated metabolic risk associated with this fat depot.
Collapse
Affiliation(s)
- Theresa Schöttl
- Molecular Nutritional Medicine, Technische Universität München, Else Kröner Fresenius Center for Nutritional Medicine, 85350 Freising, Germany
| | | | | | | | | |
Collapse
|
49
|
McCourt AC, Parker J, Silajdžić E, Haider S, Sethi H, Tabrizi SJ, Warner TT, Björkqvist M. Analysis of White Adipose Tissue Gene Expression Reveals CREB1 Pathway Altered in Huntington's Disease. J Huntingtons Dis 2015; 4:371-82. [PMID: 26756592 DOI: 10.3233/jhd-150172] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND In addition to classical neurological symptoms, Huntington's disease (HD) is complicated by peripheral pathology and both the mutant gene and the protein are found in cells and tissues throughout the body. Despite the adipose tissue gene expression alterations described in HD mouse models, adipose tissue and its gene expression signature have not been previously explored in human HD. OBJECTIVE We investigated gene expression signatures in subcutaneous adipose tissue obtained from control subjects, premanifest HD gene carriers and manifest HD subjects with the aim to identify gene expression changes and signalling pathway alterations in adipose tissue relevant to HD. METHODS Gene expression was assessed using Affymetrix GeneChip® Human Gene 1.0 ST Array. Target genes were technically validated using real-time quantitative PCR and the expression signature was validated in an independent subject cohort. RESULTS In subcutaneous adipose tissue, more than 500 genes were significantly different in premanifest HD subjects as compared to healthy controls. Pathway analysis suggests that the differentially expressed genes found here in HD adipose tissue are involved in fatty acid metabolism pathways, angiotensin signalling pathways and immune pathways. Transcription factor analysis highlights CREB1. Using RT-qPCR, we found that MAL2, AGTR2, COBL and the transcription factor CREB1 were significantly upregulated, with CREB1 and AGT also being significantly upregulated in a separate cohort. CONCLUSIONS Distinct gene expression profiles can be seen in HD subcutaneous adipose tissue, with CREB1 highlighted as a key transcription factor.
Collapse
Affiliation(s)
- Andrew Christopher McCourt
- Brain Disease Biomarker unit, Department of Experimental Medical Science, Wallenberg Neuroscience Centre, Lund University, Lund, Sweden
| | - Jennifer Parker
- Institute of Neurology, Department of Neurodegenerative Disease, UCL, London, UK
- Magnus Life Science, Rayne Building, 5 University Street, London, UK
| | - Edina Silajdžić
- Brain Disease Biomarker unit, Department of Experimental Medical Science, Wallenberg Neuroscience Centre, Lund University, Lund, Sweden
- Faculty of Life Sciences, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Salman Haider
- Institute of Neurology, Department of Neurodegenerative Disease, UCL, London, UK
| | - Huma Sethi
- Victor Horsley Department of Neurosurgery, The National Hospital for Neurology and Neurosurgery, London, UK
| | - Sarah J Tabrizi
- Institute of Neurology, Department of Neurodegenerative Disease, UCL, London, UK
| | - Thomas T Warner
- Reta Lila Weston Institute, UCL Institute of Neurology, London, UK
| | - Maria Björkqvist
- Brain Disease Biomarker unit, Department of Experimental Medical Science, Wallenberg Neuroscience Centre, Lund University, Lund, Sweden
| |
Collapse
|
50
|
Park JW, Jung KH, Lee JH, Quach CHT, Moon SH, Cho YS, Lee KH. 18F-FDG PET/CT monitoring of β3 agonist-stimulated brown adipocyte recruitment in white adipose tissue. J Nucl Med 2014; 56:153-8. [PMID: 25525187 DOI: 10.2967/jnumed.114.147603] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
UNLABELLED There is rising interest in recruitment of brown adipocytes into white adipose tissue (WAT) as a means to augment energy expenditure for weight reduction. We thus investigated the potential of (18)F-FDG uptake as an imaging biomarker that can monitor the process of WAT browning. METHODS C57BL/6 mice were treated daily with the β3 agonist CL316,243 (5-[(2R)-2-[[(2R)-2-(3-chlorophenyl)-2-hydroxyethyl]amino]propyl]-1,3-benzodioxole-2,2-dicarboxylic acid disodium salt), whereas controls received saline. (18)F-FDG small-animal PET/CT was serially performed at 1 h after CL316,243 injection. After sacrifice, interscapular brown adipose tissue (BAT) and WAT depots were extracted, weighed, and measured for (18)F-FDG uptake. Tissues underwent immunostaining, and UCP1 content was quantified by Western blotting. RESULTS PET/CT showed low (18)F-FDG uptake in both BAT and inguinal WAT at baseline. BAT uptake was substantially increased by a single stimulation with CL316,243. Uptake in inguinal WAT was only modestly elevated by the first stimulation uptake but gradually increased to BAT level by prolonged stimulation. Ex vivo measurements recapitulated the PET findings, and measured (18)F-FDG uptake in other WAT depots was similar to inguinal WAT. WAT browning by prolonged stimulation was confirmed by a substantial increase in uncoupling protein 1 (UCP1), cytochrome-c oxidase 4 (COX4), and PR domain containing 16 (PRDM16) staining as markers of brown adipocytes. UCP1 content, which served as a measure for extent of browning, was low in baseline inguinal WAT but linearly increased over 10 d of CL316,243 injection. Finally, image-based and ex vivo-measured (18)F-FDG uptake in inguinal WAT correlated well with UCP1 content. CONCLUSION (18)F-FDG PET/CT has the capacity to monitor brown adipocyte recruitment into WAT depots in vivo and may thus be useful for screening the efficacy of strategies to promote WAT browning.
Collapse
Affiliation(s)
- Jin Won Park
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyung-Ho Jung
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jin Hee Lee
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Cung Hoa Thien Quach
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seung-Hwan Moon
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Seok Cho
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyung-Han Lee
- Department of Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|