1
|
Janssen JAMJL. The Causal Role of Ectopic Fat Deposition in the Pathogenesis of Metabolic Syndrome. Int J Mol Sci 2024; 25:13238. [PMID: 39769002 PMCID: PMC11675790 DOI: 10.3390/ijms252413238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/03/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Consuming a "modern" Western diet and overnutrition may increase insulin secretion. Additionally, nutrition-mediated hyperinsulinemia is a major driver of ectopic fat deposition. The global prevalence of metabolic syndrome is high and growing. Within this context, people with congenital lipodystrophy often experience a severe form of metabolic syndrome. Evidence is increasingly supporting that subtle partial lipodystrophy plays an important role in the development of metabolic syndrome in the general population. In individuals in the general population with subtle partial lipodystrophy, as well as in those with congenital lipodystrophy, the subcutaneous adipose tissues are unable to accommodate surplus energy intake. In both conditions, (excess) fat is directed toward the liver, pancreas, and muscles, where it is deposited as ectopic fat, as this fat can no longer be stored in the "safe" subcutaneous fat depots. Ectopic fat depositions cause insulin resistance in the liver and muscles, as well as β-cell dysfunction in the pancreas. Support of a direct pathological role of ectopic fat deposition in this condition is further provided by the rapid normalization of hepatic insulin sensitivity and improvement in pancreatic β-cell function after marked reductions in ectopic fat depositions. Thus, ectopic fat deposition in the liver, pancreas, and muscles may play a causal role in the pathogenesis of metabolic syndrome even in the general population. As such, the prevention of ectopic fat deposition may reduce the risk of metabolic syndrome and mitigate its effects.
Collapse
Affiliation(s)
- Joseph A M J L Janssen
- Department of Internal Medicine, Erasmus Medical Center (Erasmus MC), Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
2
|
Al Harake SN, Abedin Y, Hatoum F, Nassar NZ, Ali A, Nassar A, Kanaan A, Bazzi S, Azar S, Harb F, Ghadieh HE. Involvement of a battery of investigated genes in lipid droplet pathophysiology and associated comorbidities. Adipocyte 2024; 13:2403380. [PMID: 39329369 PMCID: PMC11445895 DOI: 10.1080/21623945.2024.2403380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Lipid droplets (LDs) are highly specialized energy storage organelles involved in the maintenance of lipid homoeostasis by regulating lipid flux within white adipose tissue (WAT). The physiological function of adipocytes and LDs can be compromised by mutations in several genes, leading to NEFA-induced lipotoxicity, which ultimately manifests as metabolic complications, predominantly in the form of dyslipidemia, ectopic fat accumulation, and insulin resistance. In this review, we delineate the effects of mutations and deficiencies in genes - CIDEC, PPARG, BSCL2, AGPAT2, PLIN1, LIPE, LMNA, CAV1, CEACAM1, and INSR - involved in lipid droplet metabolism and their associated pathophysiological impairments, highlighting their roles in the development of lipodystrophies and metabolic dysfunction.
Collapse
Affiliation(s)
- Sami N. Al Harake
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Yasamin Abedin
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Fatema Hatoum
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Nour Zahraa Nassar
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Ali Ali
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Aline Nassar
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Amjad Kanaan
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Samer Bazzi
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Sami Azar
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Frederic Harb
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Hilda E. Ghadieh
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| |
Collapse
|
3
|
Tol MJ, Shimanaka Y, Bedard AH, Sapia J, Cui L, Colaço-Gaspar M, Hofer P, Ferrari A, Qian K, Kennelly JP, Lee SD, Gao Y, Xiao X, Gao J, Mack JJ, Weston TA, Pan C, Lusis AJ, Williams KJ, Su B, Pike DP, Reed A, Milosevich N, Cravatt BF, Arita M, Young SG, Ford DA, Zechner R, Vanni S, Tontonoz P. Dietary control of peripheral adipose storage capacity through membrane lipid remodelling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620374. [PMID: 39554041 PMCID: PMC11565995 DOI: 10.1101/2024.10.25.620374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Complex genetic and dietary cues contribute to the development of obesity, but how these are integrated on a molecular level is incompletely understood. Here, we show that PPARγ supports hypertrophic expansion of adipose tissue via transcriptional control of LPCAT3, a membrane-bound O-acyltransferase that enriches diet-derived omega-6 ( n -6) polyunsaturated fatty acids (PUFAs) in the phospholipidome. In high-fat diet-fed mice, lowering membrane n -6 PUFA levels by adipocyte-specific Lpcat3 knockout ( Lpcat3 AKO ) or by dietary lipid manipulation leads to dysfunctional triglyceride (TG) storage, ectopic fat deposition and insulin resistance. Aberrant lipolysis of stored TGs in Lpcat3 AKO adipose tissues instigates a non-canonical adaptive response that engages a futile lipid cycle to increase energy expenditure and limit further body weight gain. Mechanistically, we find that adipocyte LPCAT3 activity promotes TG storage by selectively enriching n -6 arachidonoyl-phosphatidylethanolamine at the ER-lipid droplet interface, which in turn favours the budding of large droplets that exhibit greater resistance to ATGL-dependent hydrolysis. Thus, our study highlights the PPARγ-LPCAT3 pathway as a molecular link between dietary n -6 PUFA intake, adipose expandability and systemic energy balance.
Collapse
|
4
|
Wang G, Muñoz-Rojas AR, Spallanzani RG, Franklin RA, Benoist C, Mathis D. Adipose-tissue Treg cells restrain differentiation of stromal adipocyte precursors to promote insulin sensitivity and metabolic homeostasis. Immunity 2024; 57:1345-1359.e5. [PMID: 38692280 PMCID: PMC11188921 DOI: 10.1016/j.immuni.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 02/08/2024] [Accepted: 04/04/2024] [Indexed: 05/03/2024]
Abstract
Regulatory T (Treg) cells in epidydimal visceral adipose tissue (eVAT) of lean mice and humans regulate metabolic homeostasis. We found that constitutive or punctual depletion of eVAT-Treg cells reined in the differentiation of stromal adipocyte precursors. Co-culture of these precursors with conditional medium from eVAT-Treg cells limited their differentiation in vitro, suggesting a direct effect. Transcriptional comparison of adipocyte precursors, matured in the presence or absence of the eVAT-Treg-conditioned medium, identified the oncostatin-M (OSM) signaling pathway as a key distinction. Addition of OSM to in vitro cultures blocked the differentiation of adipocyte precursors, while co-addition of anti-OSM antibodies reversed the ability of the eVAT-Treg-conditioned medium to inhibit in vitro adipogenesis. Genetic depletion of OSM (specifically in Treg) cells or of the OSM receptor (specifically on stromal cells) strongly impaired insulin sensitivity and related metabolic indices. Thus, Treg-cell-mediated control of local progenitor cells maintains adipose tissue and metabolic homeostasis, a regulatory axis seemingly conserved in humans.
Collapse
Affiliation(s)
- Gang Wang
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | | | | | - Ruth A Franklin
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | | | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Cobelo-Gómez S, Sánchez-Iglesias S, Fernández-Pombo A, Araújo-Vilar D. Effect of β-Estradiol on Adipogenesis in a 3T3-L1 Cell Model of Prelamin A Accumulation. Int J Mol Sci 2024; 25:1282. [PMID: 38279282 PMCID: PMC10816192 DOI: 10.3390/ijms25021282] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
The accumulation of farnesylated prelamin A has been suggested as one of the mechanisms responsible for the loss of fat in type 2 familial partial lipodystrophy due to variants in the LMNA gene. In this rare disease, fat loss appears in women after puberty, affecting sex-hormone-dependent anatomical areas. This study investigated the impact of 17-β-estradiol on adipogenesis in murine preadipocytes subjected to a pharmacologically induced accumulation of farnesylated and non-farnesylated prelamin A. To induce the accumulation of non-farnesylated or farnesylated prelamin A, 3T3-L1 cells were treated with the farnesyltransferase inhibitor 277 or the methyltransferase inhibitor N-acetyl-S-farnesyl-l-cysteine methylester. Subsequently, the cells were induced to undergo adipocyte differentiation in the presence or absence of 17-β-estradiol. Prelamin A accumulation was assessed through immunofluorescence, while real-time PCR and Western blot techniques were used to quantify several adipogenic genes and evaluate protein levels, respectively. The results showed that 17-β-estradiol increased adipogenesis, although the combination of this hormone plus farnesylated prelamin A led to a reduction in the number of mature adipocytes and the expression of the different genes involved in adipogenesis. In conclusion, the influence of farnesylated prelamin A accumulation on adipogenesis manifested only in the presence of estradiol. These in vitro findings suggest a potential mechanism that could explain the characteristic phenotype in women suffering type 2 familial partial lipodystrophy.
Collapse
Affiliation(s)
| | | | | | - David Araújo-Vilar
- Thyroid and Metabolic Diseases Unit (U.E.T.eM.), Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS)-IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (S.C.-G.); (S.S.-I.); (A.F.-P.)
| |
Collapse
|
6
|
García-López MÁ, Mora A, Corrales P, Pons T, Sánchez de Diego A, Talavera Gutiérrez A, van Wely KHM, Medina-Gómez G, Sabio G, Martínez-A C, Fischer T. DIDO is necessary for the adipogenesis that promotes diet-induced obesity. Proc Natl Acad Sci U S A 2024; 121:e2300096121. [PMID: 38194457 PMCID: PMC10801893 DOI: 10.1073/pnas.2300096121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 10/24/2023] [Indexed: 01/11/2024] Open
Abstract
The prevalence of overweight and obesity continues to rise in the population worldwide. Because it is an important predisposing factor for cancer, cardiovascular diseases, diabetes mellitus, and COVID-19, obesity reduces life expectancy. Adipose tissue (AT), the main fat storage organ with endocrine capacity, plays fundamental roles in systemic metabolism and obesity-related diseases. Dysfunctional AT can induce excess or reduced body fat (lipodystrophy). Dido1 is a marker gene for stemness; gene-targeting experiments compromised several functions ranging from cell division to embryonic stem cell differentiation, both in vivo and in vitro. We report that mutant mice lacking the DIDO N terminus show a lean phenotype. This consists of reduced AT and hypolipidemia, even when mice are fed a high-nutrient diet. DIDO mutation caused hypothermia due to lipoatrophy of white adipose tissue (WAT) and dermal fat thinning. Deep sequencing of the epididymal white fat (Epi WAT) transcriptome supported Dido1 control of the cellular lipid metabolic process. We found that, by controlling the expression of transcription factors such as C/EBPα or PPARγ, Dido1 is necessary for adipocyte differentiation, and that restoring their expression reestablished adipogenesis capacity in Dido1 mutants. Our model differs from other lipodystrophic mice and could constitute a new system for the development of therapeutic intervention in obesity.
Collapse
Affiliation(s)
- María Ángeles García-López
- Department of Immunology and Oncology, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas Campus, Universidad Autónoma de Madrid, Madrid28049, Spain
| | - Alfonso Mora
- Centro Nacional de Investigaciones Cardiovasculares, Madrid28029, Spain
| | - Patricia Corrales
- Department of Basic Sciences of Health, Area of Biochemistry and Molecular Biology, Universidad Rey Juan Carlos, Alcorcon28922, Spain
| | - Tirso Pons
- Department of Immunology and Oncology, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas Campus, Universidad Autónoma de Madrid, Madrid28049, Spain
| | - Ainhoa Sánchez de Diego
- Department of Immunology and Oncology, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas Campus, Universidad Autónoma de Madrid, Madrid28049, Spain
| | - Amaia Talavera Gutiérrez
- Department of Immunology and Oncology, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas Campus, Universidad Autónoma de Madrid, Madrid28049, Spain
| | - Karel H. M. van Wely
- Department of Immunology and Oncology, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas Campus, Universidad Autónoma de Madrid, Madrid28049, Spain
| | - Gema Medina-Gómez
- Department of Basic Sciences of Health, Area of Biochemistry and Molecular Biology, Universidad Rey Juan Carlos, Alcorcon28922, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares, Madrid28029, Spain
| | - Carlos Martínez-A
- Department of Immunology and Oncology, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas Campus, Universidad Autónoma de Madrid, Madrid28049, Spain
| | - Thierry Fischer
- Department of Immunology and Oncology, Centro Nacional de Biotecnología-Consejo Superior de Investigaciones Científicas Campus, Universidad Autónoma de Madrid, Madrid28049, Spain
| |
Collapse
|
7
|
Ely EV, Kapinski AT, Paradi SG, Tang R, Guilak F, Collins KH. Designer Fat Cells: Adipogenic Differentiation of CRISPR-Cas9 Genome-Engineered Induced Pluripotent Stem Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.26.564206. [PMID: 37961399 PMCID: PMC10634849 DOI: 10.1101/2023.10.26.564206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Adipose tissue is an active endocrine organ that can signal bidirectionally to many tissues and organ systems in the body. With obesity, adipose tissue is a source of low-level inflammation that contributes to various co-morbidities and damage to downstream effector tissues. The ability to synthesize genetically engineered adipose tissue could have critical applications in studying adipokine signaling and the use of adipose tissue for novel therapeutic strategies. This study aimed to develop a method for non-viral adipogenic differentiation of genome-edited murine induced pluripotent stem cells (iPSCs) and to test the ability of such cells to engraft in mice in vivo . Designer adipocytes were created from iPSCs, which can be readily genetically engineered using CRISPR-Cas9 to knock out or insert individual genes of interest. As a model system for adipocyte-based drug delivery, an existing iPSC cell line that transcribes interleukin 1 receptor antagonist under the endogenous macrophage chemoattractant protein-1 promoter was tested for adipogenic capabilities under these same differentiation conditions. To understand the role of various adipocyte subtypes and their impact on health and disease, an efficient method was devised for inducing browning and whitening of IPSC-derived adipocytes in culture. Finally, to study the downstream effects of designer adipocytes in vivo , we transplanted the designer adipocytes into fat-free lipodystrophic mice as a model system for studying adipose signaling in different models of disease or repair. This novel translational tissue engineering and regenerative medicine platform provides an innovative approach to studying the role of adipose interorgan communication in various conditions.
Collapse
|
8
|
Chitraju C, Fischer AW, Ambaw YA, Wang K, Yuan B, Hui S, Walther TC, Farese RV. Mice lacking triglyceride synthesis enzymes in adipose tissue are resistant to diet-induced obesity. eLife 2023; 12:RP88049. [PMID: 37782317 PMCID: PMC10545428 DOI: 10.7554/elife.88049] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023] Open
Abstract
Triglycerides (TGs) in adipocytes provide the major stores of metabolic energy in the body. Optimal amounts of TG stores are desirable as insufficient capacity to store TG, as in lipodystrophy, or exceeding the capacity for storage, as in obesity, results in metabolic disease. We hypothesized that mice lacking TG storage in adipocytes would result in excess TG storage in cell types other than adipocytes and severe lipotoxicity accompanied by metabolic disease. To test this hypothesis, we selectively deleted both TG synthesis enzymes, DGAT1 and DGAT2, in adipocytes (ADGAT DKO mice). As expected with depleted energy stores, ADGAT DKO mice did not tolerate fasting well and, with prolonged fasting, entered torpor. However, ADGAT DKO mice were unexpectedly otherwise metabolically healthy and did not accumulate TGs ectopically or develop associated metabolic perturbations, even when fed a high-fat diet. The favorable metabolic phenotype resulted from activation of energy expenditure, in part via BAT (brown adipose tissue) activation and beiging of white adipose tissue. Thus, the ADGAT DKO mice provide a fascinating new model to study the coupling of metabolic energy storage to energy expenditure.
Collapse
Affiliation(s)
- Chandramohan Chitraju
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public HealthBostonUnited States
- Department of Cell Biology, Harvard Medical SchoolBostonUnited States
| | - Alexander W Fischer
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public HealthBostonUnited States
- Department of Cell Biology, Harvard Medical SchoolBostonUnited States
| | - Yohannes A Ambaw
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public HealthBostonUnited States
- Department of Cell Biology, Harvard Medical SchoolBostonUnited States
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| | - Kun Wang
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public HealthBostonUnited States
- Department of Cell Biology, Harvard Medical SchoolBostonUnited States
| | - Bo Yuan
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public HealthBostonUnited States
| | - Sheng Hui
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public HealthBostonUnited States
| | - Tobias C Walther
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public HealthBostonUnited States
- Department of Cell Biology, Harvard Medical SchoolBostonUnited States
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
- Howard Hughes Medical InstituteBostonUnited States
| | - Robert V Farese
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public HealthBostonUnited States
- Department of Cell Biology, Harvard Medical SchoolBostonUnited States
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
| |
Collapse
|
9
|
Carpéné C, Stiliyanov Atanasov K, Les F, Mercader Barcelo J. Hyperglycemia and reduced adiposity of streptozotocin-induced diabetic mice are not alleviated by oral benzylamine supplementation. World J Diabetes 2022; 13:752-764. [PMID: 36188146 PMCID: PMC9521444 DOI: 10.4239/wjd.v13.i9.752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/13/2022] [Accepted: 08/22/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Benzylamine (Bza) oral administration delays the onset of hyperglycemia in insulin-resistant db-/- mice; a genetic model of obesity and type 2 diabetes.
AIM To extend the antihyperglycemic properties of oral benzylamine to a model of insulin-deficient type 1 diabetes.
METHODS Male Swiss mice were rendered diabetic by streptozotocin treatment (STZ) and divided in two groups: one received 0.5% Bza as drinking solution for 24 d (STZ Bza-drinking) while the other was drinking water ad libitum. Similar groups were constituted in age-matched, nondiabetic mice. Food intake, liquid intake, body weight gain and nonfasting blood glucose levels were followed during treatment. At the end of treatment, fasted glycemia, liver and white adipose tissue (WAT) mass were measured, while glucose uptake assays were performed in adipocytes.
RESULTS STZ diabetic mice presented typical features of insulin-deficient diabetes: reduced body mass and increased blood glucose levels. These altered parameters were not normalized in the Bza-drinking group in spite of restored food and water intake. Bza consumption could not reverse the severe fat depot atrophy of STZ diabetic mice. In the nondiabetic mice, no difference was found between control and Bza-drinking mice for any parameter. In isolated adipocytes, hexose uptake was partially activated by 0.1 mmol/L Bza in a manner that was obliterated in vitro by the amine oxidase inhibitor phenelzine and that remained unchanged after Bza supplementation. Oxidation of 0.1 mmol/L Bza in WAT was lower in STZ diabetic than in normoglycemic mice.
CONCLUSION Bza supplementation could not normalize the altered glucose handling of STZ diabetic mice with severe WAT atrophy. Consequently, its antidiabetic potential in obese and diabetic rodents does not apply to lipoatrophic type 1 diabetic mice.
Collapse
Affiliation(s)
- Christian Carpéné
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1297, Toulouse 31432, France
| | - Kristiyan Stiliyanov Atanasov
- Molecular Biology and One Health research group, Department of Fundamental Biology and Health Sciences, University of the Balearic Islands, Palma 07122, Spain
| | - Francisco Les
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge, Villanueva de Gállego, Zaragoza 50830, Spain
| | - Josep Mercader Barcelo
- Molecular Biology and One Health research group, Department of Fundamental Biology and Health Sciences, University of the Balearic Islands, Palma 07122, Spain
| |
Collapse
|
10
|
Akbari P, Sosina OA, Bovijn J, Landheer K, Nielsen JB, Kim M, Aykul S, De T, Haas ME, Hindy G, Lin N, Dinsmore IR, Luo JZ, Hectors S, Geraghty B, Germino M, Panagis L, Parasoglou P, Walls JR, Halasz G, Atwal GS, Jones M, LeBlanc MG, Still CD, Carey DJ, Giontella A, Orho-Melander M, Berumen J, Kuri-Morales P, Alegre-Díaz J, Torres JM, Emberson JR, Collins R, Rader DJ, Zambrowicz B, Murphy AJ, Balasubramanian S, Overton JD, Reid JG, Shuldiner AR, Cantor M, Abecasis GR, Ferreira MAR, Sleeman MW, Gusarova V, Altarejos J, Harris C, Economides AN, Idone V, Karalis K, Della Gatta G, Mirshahi T, Yancopoulos GD, Melander O, Marchini J, Tapia-Conyer R, Locke AE, Baras A, Verweij N, Lotta LA. Multiancestry exome sequencing reveals INHBE mutations associated with favorable fat distribution and protection from diabetes. Nat Commun 2022; 13:4844. [PMID: 35999217 PMCID: PMC9399235 DOI: 10.1038/s41467-022-32398-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 07/28/2022] [Indexed: 12/13/2022] Open
Abstract
Body fat distribution is a major, heritable risk factor for cardiometabolic disease, independent of overall adiposity. Using exome-sequencing in 618,375 individuals (including 160,058 non-Europeans) from the UK, Sweden and Mexico, we identify 16 genes associated with fat distribution at exome-wide significance. We show 6-fold larger effect for fat-distribution associated rare coding variants compared with fine-mapped common alleles, enrichment for genes expressed in adipose tissue and causal genes for partial lipodystrophies, and evidence of sex-dimorphism. We describe an association with favorable fat distribution (p = 1.8 × 10-09), favorable metabolic profile and protection from type 2 diabetes (~28% lower odds; p = 0.004) for heterozygous protein-truncating mutations in INHBE, which encodes a circulating growth factor of the activin family, highly and specifically expressed in hepatocytes. Our results suggest that inhibin βE is a liver-expressed negative regulator of adipose storage whose blockade may be beneficial in fat distribution-associated metabolic disease.
Collapse
Affiliation(s)
- Parsa Akbari
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Olukayode A. Sosina
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Jonas Bovijn
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Karl Landheer
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Jonas B. Nielsen
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Minhee Kim
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Senem Aykul
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Tanima De
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Mary E. Haas
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - George Hindy
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Nan Lin
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Ian R. Dinsmore
- grid.280776.c0000 0004 0394 1447Department of Molecular and Functional Genomics, Geisinger Health System, Danville, PA USA
| | - Jonathan Z. Luo
- grid.280776.c0000 0004 0394 1447Department of Molecular and Functional Genomics, Geisinger Health System, Danville, PA USA
| | - Stefanie Hectors
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Benjamin Geraghty
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Mary Germino
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Lampros Panagis
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Prodromos Parasoglou
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Johnathon R. Walls
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Gabor Halasz
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Gurinder S. Atwal
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | | | | | - Marcus Jones
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Michelle G. LeBlanc
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Christopher D. Still
- grid.280776.c0000 0004 0394 1447Geisinger Obesity Institute, Geisinger Health System, Danville, PA USA
| | - David J. Carey
- grid.280776.c0000 0004 0394 1447Geisinger Obesity Institute, Geisinger Health System, Danville, PA USA
| | - Alice Giontella
- grid.4514.40000 0001 0930 2361Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden ,grid.5611.30000 0004 1763 1124Department of Medicine, University of Verona, Verona, Italy
| | - Marju Orho-Melander
- grid.4514.40000 0001 0930 2361Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Jaime Berumen
- grid.9486.30000 0001 2159 0001Unidad de Medicina Experimental de la Facultad de Medicina de la Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Pablo Kuri-Morales
- grid.9486.30000 0001 2159 0001Unidad de Medicina Experimental de la Facultad de Medicina de la Universidad Nacional Autónoma de México, Mexico City, Mexico ,grid.419886.a0000 0001 2203 4701Instituto Tecnológico y de Estudios Superiores de Monterrey, Monterrey, Mexico
| | - Jesus Alegre-Díaz
- grid.9486.30000 0001 2159 0001Unidad de Medicina Experimental de la Facultad de Medicina de la Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jason M. Torres
- grid.4991.50000 0004 1936 8948MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK ,grid.4991.50000 0004 1936 8948Clinical Trial Service Unit & Epidemiological Studies Unit Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Jonathan R. Emberson
- grid.4991.50000 0004 1936 8948MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK ,grid.4991.50000 0004 1936 8948Clinical Trial Service Unit & Epidemiological Studies Unit Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Rory Collins
- grid.4991.50000 0004 1936 8948Clinical Trial Service Unit & Epidemiological Studies Unit Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Daniel J. Rader
- grid.25879.310000 0004 1936 8972Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Brian Zambrowicz
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Andrew J. Murphy
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Suganthi Balasubramanian
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - John D. Overton
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Jeffrey G. Reid
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Alan R. Shuldiner
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Michael Cantor
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Goncalo R. Abecasis
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Manuel A. R. Ferreira
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Mark W. Sleeman
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Viktoria Gusarova
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Judith Altarejos
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Charles Harris
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Aris N. Economides
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA ,grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Vincent Idone
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Katia Karalis
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Giusy Della Gatta
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Tooraj Mirshahi
- grid.280776.c0000 0004 0394 1447Geisinger Obesity Institute, Geisinger Health System, Danville, PA USA
| | | | - Olle Melander
- grid.4514.40000 0001 0930 2361Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden ,grid.411843.b0000 0004 0623 9987Department of Emergency and Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Jonathan Marchini
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Roberto Tapia-Conyer
- grid.419886.a0000 0001 2203 4701Instituto Tecnológico y de Estudios Superiores de Monterrey, Monterrey, Mexico
| | - Adam E. Locke
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Aris Baras
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY, USA.
| | - Niek Verweij
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Luca A. Lotta
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| |
Collapse
|
11
|
Deficiency of WTAP in hepatocytes induces lipoatrophy and non-alcoholic steatohepatitis (NASH). Nat Commun 2022; 13:4549. [PMID: 35927268 PMCID: PMC9352699 DOI: 10.1038/s41467-022-32163-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 07/18/2022] [Indexed: 01/18/2023] Open
Abstract
Ectopic lipid accumulation and inflammation are the essential signs of NASH. However, the molecular mechanisms of ectopic lipid accumulation and inflammation during NASH progression are not fully understood. Here we reported that hepatic Wilms' tumor 1-associating protein (WTAP) is a key integrative regulator of ectopic lipid accumulation and inflammation during NASH progression. Hepatic deletion of Wtap leads to NASH due to the increased lipolysis in white adipose tissue, enhanced hepatic free fatty acids uptake and induced inflammation, all of which are mediated by IGFBP1, CD36 and cytochemokines such as CCL2, respectively. WTAP binds to specific DNA motifs which are enriched in the promoters and suppresses gene expression (e.g., Igfbp1, Cd36 and Ccl2) with the involvement of HDAC1. In NASH, WTAP is tranlocated from nucleus to cytosol, which is related to CDK9-mediated phosphorylation. These data uncover a mechanism by which hepatic WTAP regulates ectopic lipid accumulation and inflammation during NASH progression. Ectopic lipid accumulation and inflammation are the essential signs of NASH. Here, the authors show that hepatic WTAP is a key integrative repressor of ectopic lipid accumulation and inflammation during NASH progression, and hepatic deletion of Wtap promotes both of them, leading to NASH
Collapse
|
12
|
Nichols K, Yiannikouris F. The Role of (Pro)Renin Receptor in the Metabolic Syndrome. Curr Hypertens Rev 2022; 18:117-124. [PMID: 35170416 DOI: 10.2174/1573402118666220216104816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/29/2021] [Accepted: 01/06/2022] [Indexed: 01/27/2023]
Abstract
The prorenin receptor (PRR) is a complex multi-functional single transmembrane protein receptor that is ubiquitously expressed in organs and tissues throughout the body. PRR is involved in different cellular mechanisms that comprise the generation of Angiotensin II, the activation of Wnt/β-catenin signaling, the stimulation of ERK 1/2 pathway, and the proper functioning of the vacuolar H+-ATPase. Evidence supports the role of PRR and its soluble form, sPRR, in the classical features of the metabolic syndrome, including obesity, hypertension, diabetes, and disruption of lipid homeostasis. This review summarizes our current knowledge and highlights new advances in the pathophysiological function of PRR and sPRR in adipogenesis, adipocyte differentiation, lipolysis, glucose and insulin resistance, lipid homeostasis, energy metabolism, and blood pressure regulation.
Collapse
Affiliation(s)
- Kellea Nichols
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Frederique Yiannikouris
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
13
|
Rutkowska L, Salachna D, Lewandowski K, Lewiński A, Gach A. Familial Partial Lipodystrophy-Literature Review and Report of a Novel Variant in PPARG Expanding the Spectrum of Disease-Causing Alterations in FPLD3. Diagnostics (Basel) 2022; 12:1122. [PMID: 35626278 PMCID: PMC9139680 DOI: 10.3390/diagnostics12051122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 11/16/2022] Open
Abstract
Familial partial lipodystrophy (FPLD) is a rare genetic disorder characterized by the selective loss of adipose tissue. Its estimated prevalence is as low as 1 in 1 million. The deficiency of metabolically active adipose tissue is closely linked with a wide range of metabolic complications, such as insulin resistance, lipoatrophic diabetes, dyslipidemia with severe hypertriglyceridemia, hypertension or hepatic steatosis. Moreover, female patients often develop hyperandrogenism, hirsutism, polycystic ovaries and infertility. The two most common types are FPLD type 2 and 3. Variants within LMNA and PPARG genes account for more than 50% of all reported FPLD cases. Because of its high heterogeneity and rarity, lipodystrophy can be easily unrecognized or misdiagnosed. To determine the genetic background of FPLD in a symptomatic woman and her close family, an NGS custom panel was used to sequence LMNA and PPARG genes. The affected patient presented fat deposits in the face, neck and trunk, with fat loss combined with muscular hypertrophy in the lower extremities and hirsutism, all features first manifesting at puberty. Her clinical presentation included metabolic disturbances, including hypercholesterolemia with severe hypertriglyceridemia, diabetes mellitus and hepatic steatosis. This together with her typical fat distribution and physical features raised a suspicion of FPLD. NGS analysis revealed the presence of missense heterozygous variant c.443G>A in exon 4 of PPARG gene, causing glycine to glutamic acid substitution at amino acid position 148, p.(Gly148Glu). The variant was also found in the patient’s mother and son. The variant was not previously reported in any public database. Based on computational analysis, crucial variant localization within DNA-binding domain of PPARγ, available literature data and the variant cosegregation in the patient’s family, novel c.443G>A variant was suspected to be causative. Functional testing is needed to confirm the pathogenicity of the novel variant. Inherited lipodystrophy syndromes represent a heterogenous group of metabolic disorders, whose background often remains unclear. A better understating of the genetic basis would allow earlier diagnosis and targeted treatment implementation.
Collapse
Affiliation(s)
- Lena Rutkowska
- Department of Genetics, Polish Mother’s Memorial Hospital-Research Institute, 93-338 Lodz, Poland;
| | - Dominik Salachna
- Department of Genetics, Polish Mother’s Memorial Hospital-Research Institute, 93-338 Lodz, Poland;
| | - Krzysztof Lewandowski
- Department of Endocrinology and Metabolic Diseases, Medical University of Lodz, 90-419 Lodz, Poland
- Department of Endocrinology and Metabolic Diseases, Polish Mother’s Memorial Hospital-Research Institute, 93-338 Lodz, Poland
| | - Andrzej Lewiński
- Department of Endocrinology and Metabolic Diseases, Medical University of Lodz, 90-419 Lodz, Poland
- Department of Endocrinology and Metabolic Diseases, Polish Mother’s Memorial Hospital-Research Institute, 93-338 Lodz, Poland
| | - Agnieszka Gach
- Department of Genetics, Polish Mother’s Memorial Hospital-Research Institute, 93-338 Lodz, Poland;
| |
Collapse
|
14
|
Mainieri F, Tagi VM, Chiarelli F. Treatment Options for Lipodystrophy in Children. Front Endocrinol (Lausanne) 2022; 13:879979. [PMID: 35600578 PMCID: PMC9114741 DOI: 10.3389/fendo.2022.879979] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 03/31/2022] [Indexed: 11/25/2022] Open
Abstract
Lipodystrophy includes a heterogeneous group of rare diseases characterized by different amounts of adipose tissue loss and several metabolic complications, including hypertriglyceridemia, steatohepatitis and particularly insulin resistance, that may lead to severe morbidity and, sometimes, mortality. Therefore, therapy for lipodystrophy primarily consists of a conventional approach that involves standard treatments of metabolic abnormalities. Given the evidence of leptin deficiency in lipodystrophy syndromes, leptin replacement therapy has been considered as a treatment option. Long-term studies on the use of therapy with a methionylated analog of human leptin, metreleptin, first on animals and subsequently on human patients, demonstrated enormous improvements of patients' clinical features and metabolic conditions. Recently, metreleptin was approved by Food and Drug Administration (FDA) for the treatment of generalized lipodystrophy and by European Medicines Agency (EMA) for the treatment of both generalized and partial lipodystrophy. However, further research is being conducted for new and different therapeutic agents, especially helpful for the treatment of patients with partial lipodystrophy, as some of them do not have access to metreleptin therapy or show poor response.
Collapse
|
15
|
Lacroix M, Linares LK, Rueda-Rincon N, Bloch K, Di Michele M, De Blasio C, Fau C, Gayte L, Blanchet E, Mairal A, Derua R, Cardona F, Beuzelin D, Annicotte JS, Pirot N, Torro A, Tinahones FJ, Bernex F, Bertrand-Michel J, Langin D, Fajas L, Swinnen JV, Le Cam L. The multifunctional protein E4F1 links P53 to lipid metabolism in adipocytes. Nat Commun 2021; 12:7037. [PMID: 34857760 PMCID: PMC8639890 DOI: 10.1038/s41467-021-27307-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 11/12/2021] [Indexed: 01/20/2023] Open
Abstract
Growing evidence supports the importance of the p53 tumor suppressor in metabolism but the mechanisms underlying p53-mediated control of metabolism remain poorly understood. Here, we identify the multifunctional E4F1 protein as a key regulator of p53 metabolic functions in adipocytes. While E4F1 expression is upregulated during obesity, E4f1 inactivation in mouse adipose tissue results in a lean phenotype associated with insulin resistance and protection against induced obesity. Adipocytes lacking E4F1 activate a p53-dependent transcriptional program involved in lipid metabolism. The direct interaction between E4F1 and p53 and their co-recruitment to the Steaoryl-CoA Desaturase-1 locus play an important role to regulate monounsaturated fatty acids synthesis in adipocytes. Consistent with the role of this E4F1-p53-Steaoryl-CoA Desaturase-1 axis in adipocytes, p53 inactivation or diet complementation with oleate partly restore adiposity and improve insulin sensitivity in E4F1-deficient mice. Altogether, our findings identify a crosstalk between E4F1 and p53 in the control of lipid metabolism in adipocytes that is relevant to obesity and insulin resistance.
Collapse
Affiliation(s)
- Matthieu Lacroix
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Univ Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France.,Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - Laetitia K Linares
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Univ Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France.,Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - Natalia Rueda-Rincon
- KU Leuven-University of Leuven, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven, Belgium
| | - Katarzyna Bloch
- KU Leuven-University of Leuven, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven, Belgium
| | - Michela Di Michele
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Univ Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France.,Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - Carlo De Blasio
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Univ Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France.,Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - Caroline Fau
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Univ Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France.,Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - Laurie Gayte
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Univ Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France.,Equipe labélisée Ligue Contre le Cancer, Paris, France
| | - Emilie Blanchet
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Univ Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Aline Mairal
- I2MC, Institute of Metabolic and Cardiovascular Diseases, Université de Toulouse, INSERM, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Rita Derua
- KU Leuven-University of Leuven, Department of Cellular and Molecular Medicine, Leuven, Belgium
| | - Fernando Cardona
- Department of Surgical Specialties, Biochemistry and Immunology School of Medicine, University of Malaga, Malaga, Spain
| | - Diane Beuzelin
- I2MC, Institute of Metabolic and Cardiovascular Diseases, Université de Toulouse, INSERM, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Jean-Sebastien Annicotte
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, CNRS, U1283 - UMR 8199 - EGID, Lille, France
| | - Nelly Pirot
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Univ Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France.,BioCampus, RHEM, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Adeline Torro
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Univ Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Francisco J Tinahones
- CIBER of Physiopathology, Obesity and Nutrition (CIBEROBN), Málaga, Spain; Unidad de Gestion Clinica de Endocrinologia y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Clinico Virgen de la Victoria, Málaga, Spain
| | - Florence Bernex
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Univ Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France.,BioCampus, RHEM, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Justine Bertrand-Michel
- I2MC, Institute of Metabolic and Cardiovascular Diseases, Université de Toulouse, INSERM, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Dominique Langin
- I2MC, Institute of Metabolic and Cardiovascular Diseases, Université de Toulouse, INSERM, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France.,Toulouse University Hospitals, Department of Clinical Biochemistry, Toulouse, France
| | - Lluis Fajas
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Johannes V Swinnen
- KU Leuven-University of Leuven, Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven, Belgium
| | - Laurent Le Cam
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Univ Montpellier, Institut régional du Cancer de Montpellier, Montpellier, France. .,Equipe labélisée Ligue Contre le Cancer, Paris, France.
| |
Collapse
|
16
|
Hunyenyiwa T, Hendee K, Matus K, Kyi P, Mammoto T, Mammoto A. Obesity Inhibits Angiogenesis Through TWIST1-SLIT2 Signaling. Front Cell Dev Biol 2021; 9:693410. [PMID: 34660572 PMCID: PMC8511494 DOI: 10.3389/fcell.2021.693410] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 08/30/2021] [Indexed: 01/22/2023] Open
Abstract
Angiogenesis is required for functional adipose tissue maintenance, remodeling, and expansion. Physiologically balanced adipogenesis and angiogenesis are inhibited in subcutaneous adipose tissue in obese humans. However, the mechanism by which angiogenesis is inhibited in obese adipose tissue is not fully understood. Transcription factor TWIST1 controls angiogenesis and vascular function. TWIST1 expression is lower in obese human adipose tissues. Here, we have demonstrated that angiogenesis is inhibited in endothelial cells (ECs) isolated from adipose tissues of obese humans through TWIST1-SLIT2 signaling. The levels of TWIST1 and SLIT2 are lower in ECs isolated from obese human adipose tissues compared to those from lean tissues. Knockdown of TWIST1 in lean human adipose ECs decreases, while overexpression of TWIST1 in obese adipose ECs restores SLIT2 expression. DNA synthesis and cell migration are inhibited in obese adipose ECs and the effects are restored by TWIST1 overexpression. Obese adipose ECs also inhibit blood vessel formation in the gel subcutaneously implanted in mice, while these effects are restored when gels are mixed with SLIT2 or supplemented with ECs overexpressing TWIST1. These findings suggest that obesity impairs adipose tissue angiogenesis through TWIST1-SLIT2 signaling.
Collapse
Affiliation(s)
- Tendai Hunyenyiwa
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Kathryn Hendee
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Kienna Matus
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Priscilla Kyi
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Tadanori Mammoto
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Akiko Mammoto
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
17
|
Independent Dose-Response Associations between Fetuin-A and Lean Nonalcoholic Fatty Liver Disease. Nutrients 2021; 13:nu13092928. [PMID: 34578806 PMCID: PMC8468081 DOI: 10.3390/nu13092928] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/23/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022] Open
Abstract
Patients with lean NAFLD make up an increasing subset of liver disease patients. The association between lean NAFLD and feutin-A, which serves as a hepatokine and adipokine, has never been examined. Our study aimed to explore the association of serum fetuin-A among lean and non-lean patients. The study comprised 606 adults from the community, stratified into lean or non-lean (BMI </≥ 24 kg/m2) and NAFLD or non-NAFLD (scoring of ultrasonographic fatty liver indicator, US-FLI ≥ 2/< 2). Multivariate logistic regression analyses were performed to estimate the odds ratio of having NAFLD among the tertiles of fetuin-A after adjustment. The least square means were computed by general linear models to estimate marginal means of the serum fetuin-A concentrations in relation to the NAFLD groups. The odds ratio (OR) of having NAFLD for the highest versus the lowest tertile of fetuin-A was 2.62 (95% CI: 1.72–3.98; p for trend < 0.001). Stratifying by BMI, the OR of having lean NAFLD for the highest versus the lowest tertile of fetuin-A was 2.09 (95% CI: 1.09–3.98; p for trend 0.026), while non-lean NAFLD had no significant association with the fetuin-A gradient after adjustments. Fetuin-A was positively associated with lean NAFLD after adjusting for central obesity and insulin resistance.
Collapse
|
18
|
Nipping Adipocyte Inflammation in the Bud. ACTA ACUST UNITED AC 2021; 3. [PMID: 33732506 PMCID: PMC7963359 DOI: 10.20900/immunometab20210012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Adipose tissue inflammation continues to represent a significant area of research in immunometabolism. We have identified a transcription factor, EBF1, which crucially regulates the expression of numerous inflammatory loci in adipocytes. However, EBF1 appears to do so without physically binding to these inflammatory genes. Our research is currently focused on understanding this discrepancy, and we believe that future findings could pave the road for drug development aimed to block adipose inflammation at its source.
Collapse
|
19
|
Alvarez-Guaita A, Patel S, Lim K, Haider A, Dong L, Conway OJ, Ma MKL, Chiarugi D, Saudek V, O'Rahilly S, Savage DB. Phenotypic characterization of Adig null mice suggests roles for adipogenin in the regulation of fat mass accrual and leptin secretion. Cell Rep 2021; 34:108810. [PMID: 33691105 PMCID: PMC7966854 DOI: 10.1016/j.celrep.2021.108810] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 12/23/2020] [Accepted: 02/09/2021] [Indexed: 12/25/2022] Open
Abstract
Adipogenin (Adig) is an adipocyte-enriched transmembrane protein. Its expression is induced during adipogenesis in rodent cells, and a recent genome-wide association study associated body mass index (BMI)-adjusted leptin levels with the ADIG locus. In order to begin to understand the biological function of Adig, we studied adipogenesis in Adig-deficient cultured adipocytes and phenotyped Adig null (Adig-/-) mice. Data from Adig-deficient cells suggest that Adig is required for adipogenesis. In vivo, Adig-/- mice are leaner than wild-type mice when fed a high-fat diet and when crossed with Ob/Ob hyperphagic mice. In addition to the impact on fat mass accrual, Adig deficiency also reduces fat-mass-adjusted plasma leptin levels and impairs leptin secretion from adipose explants, suggesting an additional impact on the regulation of leptin secretion.
Collapse
Affiliation(s)
- Anna Alvarez-Guaita
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, Cambridgeshire CB2 0QQ, UK
| | - Satish Patel
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, Cambridgeshire CB2 0QQ, UK
| | - Koini Lim
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, Cambridgeshire CB2 0QQ, UK
| | - Afreen Haider
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, Cambridgeshire CB2 0QQ, UK
| | - Liang Dong
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, Cambridgeshire CB2 0QQ, UK
| | - Olivia J Conway
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, Cambridgeshire CB2 0QQ, UK
| | - Marcella K L Ma
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Genomics and Transcriptomics Core, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Davide Chiarugi
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, Cambridgeshire CB2 0QQ, UK
| | - Vladimir Saudek
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, Cambridgeshire CB2 0QQ, UK
| | - Stephen O'Rahilly
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, Cambridgeshire CB2 0QQ, UK
| | - David B Savage
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, Cambridgeshire CB2 0QQ, UK.
| |
Collapse
|
20
|
Park J, Sohn JH, Han SM, Park YJ, Huh JY, Choe SS, Kim JB. Adipocytes Are the Control Tower That Manages Adipose Tissue Immunity by Regulating Lipid Metabolism. Front Immunol 2021; 11:598566. [PMID: 33584664 PMCID: PMC7876236 DOI: 10.3389/fimmu.2020.598566] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/10/2020] [Indexed: 12/14/2022] Open
Abstract
Accumulating evidence reveals that adipose tissue is an immunologically active organ that exerts multiple impacts on the regulation of systemic energy metabolism. Adipose tissue immunity is modulated by the interactions between adipocytes and various immune cells. Nevertheless, the underlying mechanisms that control inter-cellular interactions between adipocytes and immune cells in adipose tissue have not been thoroughly elucidated. Recently, it has been demonstrated that adipocytes utilize lipid metabolites as a key mediator to initiate and mediate diverse adipose tissue immune responses. Adipocytes present lipid antigens and secrete lipid metabolites to determine adipose immune tones. In addition, the interactions between adipocytes and adipose immune cells are engaged in the control of adipocyte fate and functions upon metabolic stimuli. In this review, we discuss an integrated view of how adipocytes communicate with adipose immune cells using lipid metabolites. Also, we briefly discuss the newly discovered roles of adipose stem cells in the regulation of adipose tissue immunity.
Collapse
Affiliation(s)
- Jeu Park
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Seoul National University, Seoul, South Korea.,Institute of Molecular Biology and Genetics, Seoul National University, Seoul, South Korea.,School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Jee Hyung Sohn
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Seoul National University, Seoul, South Korea.,Institute of Molecular Biology and Genetics, Seoul National University, Seoul, South Korea.,School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Sang Mun Han
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Seoul National University, Seoul, South Korea.,Institute of Molecular Biology and Genetics, Seoul National University, Seoul, South Korea.,School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Yoon Jeong Park
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Seoul National University, Seoul, South Korea.,Institute of Molecular Biology and Genetics, Seoul National University, Seoul, South Korea.,School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Jin Young Huh
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Seoul National University, Seoul, South Korea.,Institute of Molecular Biology and Genetics, Seoul National University, Seoul, South Korea.,School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Sung Sik Choe
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Seoul National University, Seoul, South Korea.,Institute of Molecular Biology and Genetics, Seoul National University, Seoul, South Korea.,School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Jae Bum Kim
- National Creative Research Initiatives Center for Adipocyte Structure and Function, Seoul National University, Seoul, South Korea.,Institute of Molecular Biology and Genetics, Seoul National University, Seoul, South Korea.,School of Biological Sciences, Seoul National University, Seoul, South Korea
| |
Collapse
|
21
|
Dean JM, He A, Tan M, Wang J, Lu D, Razani B, Lodhi IJ. MED19 Regulates Adipogenesis and Maintenance of White Adipose Tissue Mass by Mediating PPARγ-Dependent Gene Expression. Cell Rep 2020; 33:108228. [PMID: 33027649 PMCID: PMC7561447 DOI: 10.1016/j.celrep.2020.108228] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 08/11/2020] [Accepted: 09/14/2020] [Indexed: 12/13/2022] Open
Abstract
The Mediator complex relays regulatory signals from gene-specific transcription factors to the basal transcriptional machinery. However, the role of individual Mediator subunits in different tissues remains unclear. Here, we demonstrate that MED19 is essential for adipogenesis and maintenance of white adipose tissue (WAT) by mediating peroxisome proliferator-activated receptor gamma (PPARγ) transcriptional activity. MED19 knockdown blocks white adipogenesis, but not brown adipogenesis or C2C12 myoblast differentiation. Adipose-specific MED19 knockout (KO) in mice results in a striking loss of WAT, whitening of brown fat, hepatic steatosis, and insulin resistance. Inducible adipose-specific MED19 KO in adult animals also results in lipodystrophy, demonstrating its requirement for WAT maintenance. Global gene expression analysis reveals induction of genes involved in apoptosis and inflammation and impaired expression of adipose-specific genes, resulting from decreased PPARγ residency on adipocyte gene promoters and reduced association of PPARγ with RNA polymerase II. These results identify MED19 as a crucial facilitator of PPARγ-mediated gene expression in adipose tissue.
Collapse
Affiliation(s)
- John M Dean
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anyuan He
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Min Tan
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jun Wang
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dongliang Lu
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Babak Razani
- Cardiology Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Veterans Affairs St. Louis Healthcare System, John Cochran Division, St. Louis, MO 63106, USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
22
|
Liang X, Tao C, Pan J, Zhang L, Liu L, Zhao Y, Fan Y, Cao C, Liu J, Zhang J, Lam SM, Shui G, Jin W, Li W, Zhao J, Li K, Wang Y. Rnf20 deficiency in adipocyte impairs adipose tissue development and thermogenesis. Protein Cell 2020; 12:475-492. [PMID: 32797353 PMCID: PMC8160045 DOI: 10.1007/s13238-020-00770-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 07/05/2020] [Indexed: 12/14/2022] Open
Abstract
RNF20, an E3 ligase critical for monoubiquitination of histone H2B at lysine 120 (H2Bub), has been implicated in the regulation of various cellar processes; however, its physiological roles in adipocytes remain poorly characterized. Here, we report that the adipocyte-specific knockout of Rnf20 (ASKO) in mice led to progressive fat loss, organomegaly and hyperinsulinemia. Despite signs of hyperinsulinemia, normal insulin sensitivity and improved glucose tolerance were observed in the young and aged CD-fed ASKO mice. In addition, high-fat diet-fed ASKO mice developed severe liver steatosis. Moreover, we observed that the ASKO mice were extremely sensitive to a cold environment due to decreased expression levels of brown adipose tissue (BAT) selective genes, including uncoupling protein 1 (Ucp1), and impaired mitochondrial functions. Significantly decreased levels of peroxisome proliferator-activated receptor gamma (Pparγ) were observed in the gonadal white adipose tissues (gWAT) from the ASKO mice, suggesting that Rnf20 regulates adipogenesis, at least in part, through Pparγ. Rosiglitazone-treated ASKO mice exhibited increased fat mass compared to that of the non-treated ASKO mice. Collectively, our results illustrate the critical role of RNF20 in control of white and brown adipose tissue development and physiological function.
Collapse
Affiliation(s)
- Xiaojuan Liang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Cong Tao
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Jianfei Pan
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Lilan Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Lulu Liu
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
- Department of Animal Science, China Agricultural University, Beijing, 100193, China
| | - Ying Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yiping Fan
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Chunwei Cao
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiali Liu
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Jin Zhang
- College of Biological, Chemical Sciences and Engineering, Jiaxing University, Jiaxing, 314001, China
| | - Sin Man Lam
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Guanghou Shui
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wanzhu Jin
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Li
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jianguo Zhao
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Kui Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - Yanfang Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
23
|
Aprile M, Cataldi S, Perfetto C, Ambrosio MR, Italiani P, Tatè R, Blüher M, Ciccodicola A, Costa V. In-Vitro-Generated Hypertrophic-Like Adipocytes Displaying PPARG Isoforms Unbalance Recapitulate Adipocyte Dysfunctions In Vivo. Cells 2020; 9:cells9051284. [PMID: 32455814 PMCID: PMC7290899 DOI: 10.3390/cells9051284] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 12/30/2022] Open
Abstract
Reduced neo-adipogenesis and dysfunctional lipid-overloaded adipocytes are hallmarks of hypertrophic obesity linked to insulin resistance. Identifying molecular features of hypertrophic adipocytes requires appropriate in vitro models. We describe the generation of a model of human hypertrophic-like adipocytes directly comparable to normal adipose cells and the pathologic evolution toward hypertrophic state. We generate in vitro hypertrophic cells from mature adipocytes, differentiated from human mesenchymal stem cells. Combining optical, confocal, and transmission electron microscopy with mRNA/protein quantification, we characterize this cellular model, confirming specific alterations also in subcutaneous adipose tissue. Specifically, we report the generation and morphological/molecular characterization of human normal and hypertrophic-like adipocytes. The latter displays altered morphology and unbalance between canonical and dominant negative (PPARGΔ5) transcripts of PPARG, paralleled by reduced expression of PPARγ targets, including GLUT4. Furthermore, the unbalance of PPARγ isoforms associates with GLUT4 down-regulation in subcutaneous adipose tissue of individuals with overweight/obesity or impaired glucose tolerance/type 2 diabetes, but not with normal weight or glucose tolerance. In conclusion, the hypertrophic-like cells described herein are an innovative tool for studying molecular dysfunctions in hypertrophic obesity and the unbalance between PPARγ isoforms associates with down-regulation of GLUT4 and other PPARγ targets, representing a new hallmark of hypertrophic adipocytes.
Collapse
Affiliation(s)
- Marianna Aprile
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso,” CNR, Via P. Castellino 111, 80131 Naples, Italy; (S.C.); (C.P.); (R.T.); (A.C.)
- Correspondence: (M.A.); (V.C.)
| | - Simona Cataldi
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso,” CNR, Via P. Castellino 111, 80131 Naples, Italy; (S.C.); (C.P.); (R.T.); (A.C.)
| | - Caterina Perfetto
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso,” CNR, Via P. Castellino 111, 80131 Naples, Italy; (S.C.); (C.P.); (R.T.); (A.C.)
| | - Maria Rosaria Ambrosio
- Department of Translational Medicine, University of Naples “Federico II” & URT “Genomic of Diabetes,” Institute of Experimental Endocrinology and Oncology “G. Salvatore,” CNR, Via Pansini 5, 80131 Naples, Italy;
| | - Paola Italiani
- Institute of Biochemistry and Cell Biology CNR, Via P. Castellino 111, 80131 Naples, Italy;
| | - Rosarita Tatè
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso,” CNR, Via P. Castellino 111, 80131 Naples, Italy; (S.C.); (C.P.); (R.T.); (A.C.)
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, 4289 Leipzig, Germany;
| | - Alfredo Ciccodicola
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso,” CNR, Via P. Castellino 111, 80131 Naples, Italy; (S.C.); (C.P.); (R.T.); (A.C.)
- Department of Science and Technology, University of Naples “Parthenope,” 80131 Naples, Italy
| | - Valerio Costa
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso,” CNR, Via P. Castellino 111, 80131 Naples, Italy; (S.C.); (C.P.); (R.T.); (A.C.)
- Correspondence: (M.A.); (V.C.)
| |
Collapse
|
24
|
Spatiotemporal contact between peroxisomes and lipid droplets regulates fasting-induced lipolysis via PEX5. Nat Commun 2020; 11:578. [PMID: 31996685 PMCID: PMC6989686 DOI: 10.1038/s41467-019-14176-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 12/17/2019] [Indexed: 01/05/2023] Open
Abstract
Lipid droplets (LDs) are key subcellular organelles for regulating lipid metabolism. Although several subcellular organelles participate in lipid metabolism, it remains elusive whether physical contacts between subcellular organelles and LDs might be involved in lipolysis upon nutritional deprivation. Here, we demonstrate that peroxisomes and peroxisomal protein PEX5 mediate fasting-induced lipolysis by stimulating adipose triglyceride lipase (ATGL) translocation onto LDs. During fasting, physical contacts between peroxisomes and LDs are increased by KIFC3-dependent movement of peroxisomes toward LDs, which facilitates spatial translocations of ATGL onto LDs. In addition, PEX5 could escort ATGL to contact points between peroxisomes and LDs in the presence of fasting cues. Moreover, in adipocyte-specific PEX5-knockout mice, the recruitment of ATGL onto LDs was defective and fasting-induced lipolysis is attenuated. Collectively, these data suggest that physical contacts between peroxisomes and LDs are required for spatiotemporal translocation of ATGL, which is escorted by PEX5 upon fasting, to maintain energy homeostasis. Lipid droplets are organelles that regulate lipid metabolism but if organellar contacts play a role during lipolysis is unclear. Here, the authors show that peroxisomes and peroxisomal protein PEX5 play pivotal roles in the spatial and temporal regulation of fasting-induced lipolysis by translocating ATGL onto lipid droplets
Collapse
|
25
|
Matias I, Dias S, Carvalho T. Modulating the Metabolic Phenotype of Cancer Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:403-411. [PMID: 32130711 DOI: 10.1007/978-3-030-34025-4_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This chapter provides a brief overview of the methods to study and modulate the metabolic phenotype of the tumor microenvironment, including own research work to demonstrate the impact that metabolic shifts in the host have on cancer. Firstly, we briefly discuss the relevance of using animal models to address this topic, and also the importance of acknowledging that animals have diverse metabolic phenotypes according to species, and even with strain, age or sex. We also present original data to highlight the impact that changes in metabolic phenotype of the microenvironment have on tumor progression. Using an acute leukemia mouse xenograft model and high-fat diet we show that a shift in the host metabolic phenotype, induced by high-fat feeding, significantly impacts on tumor progression. The mechanism through which this occurs involves a direct effect of the increased levels of circulating lipoproteins in both tumor and non-neoplastic cells.
Collapse
Affiliation(s)
- Inês Matias
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Sérgio Dias
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - Tânia Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
26
|
Corvillo F, Akinci B. An overview of lipodystrophy and the role of the complement system. Mol Immunol 2019; 112:223-232. [PMID: 31177059 DOI: 10.1016/j.molimm.2019.05.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 05/28/2019] [Accepted: 05/29/2019] [Indexed: 12/19/2022]
Abstract
The complement system is a major component of innate immunity playing essential roles in the destruction of pathogens, the clearance of apoptotic cells and immune complexes, the enhancement of phagocytosis, inflammation, and the modulation of adaptive immune responses. During the last decades, numerous studies have shown that the complement system has key functions in the biology of certain tissues. For example, complement contributes to normal brain and embryonic development and to the homeostasis of lipid metabolism. However, the complement system is subjected to the effective balance between activation-inactivation to maintain complement homeostasis and to prevent self-injury to cells or tissues. When this control is disrupted, serious pathologies eventually develop, such as C3 glomerulopathy, autoimmune conditions and infections. Another heterogeneous group of ultra-rare diseases in which complement abnormalities have been described are the lipodystrophy syndromes. These diseases are characterized by the loss of adipose tissue throughout the entire body or partially. Complement over-activation has been reported in most of the patients with acquired partial lipodystrophy (also called Barraquer-Simons Syndrome) and in some cases of the generalized variety of the disease (Lawrence Syndrome). Even so, the mechanism through which the complement system induces adipose tissue abnormalities remains unclear. This review focuses on describing the link between the complement system and certain forms of lipodystrophy. In addition, we present an overview regarding the clinical presentation, differential diagnosis, classification, and management of patients with lipodystrophy associated with complement abnormalities.
Collapse
Affiliation(s)
- F Corvillo
- Complement Research Group, La Paz University Hospital Research Institute (IdiPAZ), La Paz University Hospital, Madrid, Spain; Center for Biomedical Network Research on Rare Diseases (CIBERER U754), Madrid, Spain.
| | - B Akinci
- Division of Endocrinology, Department of Internal Medicine, Dokuz Eylul University, Izmir, Turkey; Brehm Center for Diabetes Research, Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan, 1000 Wall Street, Room 5313, Ann Arbor, MI, 48105, USA
| |
Collapse
|
27
|
Fernandez S, Viola JM, Torres A, Wallace M, Trefely S, Zhao S, Affronti HC, Gengatharan JM, Guertin DA, Snyder NW, Metallo CM, Wellen KE. Adipocyte ACLY Facilitates Dietary Carbohydrate Handling to Maintain Metabolic Homeostasis in Females. Cell Rep 2019; 27:2772-2784.e6. [PMID: 31141698 PMCID: PMC6608748 DOI: 10.1016/j.celrep.2019.04.112] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 03/15/2019] [Accepted: 04/26/2019] [Indexed: 02/07/2023] Open
Abstract
Sugars and refined carbohydrates are major components of the modern diet. ATP-citrate lyase (ACLY) is upregulated in adipocytes in response to carbohydrate consumption and generates acetyl-coenzyme A (CoA) for both lipid synthesis and acetylation reactions. Here, we investigate the role of ACLY in the metabolic and transcriptional responses to carbohydrates in adipocytes and unexpectedly uncover a sexually dimorphic function in maintaining systemic metabolic homeostasis. When fed a high-sucrose diet, AclyFAT-/- females exhibit a lipodystrophy-like phenotype, with minimal fat accumulation, insulin resistance, and hepatic lipid accumulation, whereas AclyFAT-/- males have only mild metabolic phenotypes. We find that ACLY is crucial for nutrient-dependent carbohydrate response element-binding protein (ChREBP) activation in adipocytes and plays a key role, particularly in females, in the storage of newly synthesized fatty acids in adipose tissue. The data indicate that adipocyte ACLY is important in females for the systemic handling of dietary carbohydrates and for the preservation of metabolic homeostasis.
Collapse
Affiliation(s)
- Sully Fernandez
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - John M Viola
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - AnnMarie Torres
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Martina Wallace
- Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sophie Trefely
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; A.J. Drexel Autism Institute, Drexel University, Philadelphia, PA 19104, USA
| | - Steven Zhao
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Hayley C Affronti
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jivani M Gengatharan
- Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - David A Guertin
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Nathaniel W Snyder
- A.J. Drexel Autism Institute, Drexel University, Philadelphia, PA 19104, USA
| | - Christian M Metallo
- Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
28
|
Gatineau E, Cohn DM, Poglitsch M, Loria AS, Gong M, Yiannikouris F. Losartan prevents the elevation of blood pressure in adipose-PRR deficient female mice while elevated circulating sPRR activates the renin-angiotensin system. Am J Physiol Heart Circ Physiol 2019; 316:H506-H515. [PMID: 30550352 PMCID: PMC6734055 DOI: 10.1152/ajpheart.00473.2018] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 12/07/2018] [Accepted: 12/13/2018] [Indexed: 12/30/2022]
Abstract
Deletion of the prorenin receptor (PRR) in adipose tissue elevates systolic blood pressure (SBP) and the circulating soluble form of PRR (sPRR) in male mice fed a high-fat (HF) diet. However, sex differences in the contribution of adipose-PRR and sPRR to the regulation of the renin-angiotensin system (RAS) in key organs for blood pressure control are undefined. Therefore, we assessed blood pressure and the systemic and intrarenal RAS status in adipose-PRR knockout (KO) female mice. Blockade of RAS with losartan blunted SBP elevation in HF diet-fed adipose-PRR KO mice. ANG II levels were significantly increased in the renal cortex of HF diet-fed adipose-PRR KO female mice, but not systemically. HF diet-fed adipose-PRR KO mice exhibited higher vasopressin levels, water retention, and lower urine output than wild-type (WT) mice. The results also showed that deletion of adipose-PRR increased circulating sPRR and total hepatic sPRR contents, suggesting the liver as a major source of elevated plasma sPRR in adipose-PRR KO mice. To mimic the elevation of circulating sPRR and define the direct contribution of systemic sPRR to the regulation of the RAS and vasopressin, C57BL/6 female mice fed a standard diet were infused with recombinant sPRR. sPRR infusion increased plasma renin levels, renal and hepatic angiotensinogen expression, and vasopressin. Together, these results demonstrate that the deletion of adipose-PRR induced an elevation of SBP likely mediated by an intrarenal ANG II-dependent mechanism and that sPRR participates in RAS regulation and body fluid homeostasis via its capacity to activate the RAS and increase vasopressin levels. NEW & NOTEWORTHY The elevation of systolic blood pressure appears to be primarily mediated by cortical ANG II in high-fat diet-fed adipose-prorenin receptor knockout female mice. In addition, our data support a role for soluble prorenin receptor in renin-angiotensin system activation and vasopressin regulation.
Collapse
Affiliation(s)
- Eva Gatineau
- Department of Pharmacology and Nutritional Sciences, University of Kentucky , Lexington, Kentucky
| | - Dianne M Cohn
- Department of Pharmacology and Nutritional Sciences, University of Kentucky , Lexington, Kentucky
| | | | - Analia S Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky , Lexington, Kentucky
| | - Ming Gong
- Department of Physiology, University of Kentucky , Lexington, Kentucky
| | - Frédérique Yiannikouris
- Department of Pharmacology and Nutritional Sciences, University of Kentucky , Lexington, Kentucky
| |
Collapse
|
29
|
Akinci B, Meral R, Oral EA. Phenotypic and Genetic Characteristics of Lipodystrophy: Pathophysiology, Metabolic Abnormalities, and Comorbidities. Curr Diab Rep 2018; 18:143. [PMID: 30406415 DOI: 10.1007/s11892-018-1099-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW This article focuses on recent progress in understanding the genetics of lipodystrophy syndromes, the pathophysiology of severe metabolic abnormalities caused by these syndromes, and causes of severe morbidity and a possible signal of increased mortality associated with lipodystrophy. An updated classification scheme is also presented. RECENT FINDINGS Lipodystrophy encompasses a group of heterogeneous rare diseases characterized by generalized or partial lack of adipose tissue and associated metabolic abnormalities including altered lipid metabolism and insulin resistance. Recent advances in the field have led to the discovery of new genes associated with lipodystrophy and have also improved our understanding of adipose biology, including differentiation, lipid droplet assembly, and metabolism. Several registries have documented the natural history of the disease and the serious comorbidities that patients with lipodystrophy face. There is also evolving evidence for increased mortality rates associated with lipodystrophy. Lipodystrophy syndromes represent a challenging cluster of diseases that lead to severe insulin resistance, a myriad of metabolic abnormalities, and serious morbidity. The understanding of these syndromes is evolving in parallel with the identification of novel disease-causing mechanisms.
Collapse
Affiliation(s)
- Baris Akinci
- Brehm Center for Diabetes Research, Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan, 1000 Wall Street, Room 5313, Ann Arbor, MI, 48105, USA
- Division of Endocrinology, Department of Internal Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Rasimcan Meral
- Brehm Center for Diabetes Research, Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan, 1000 Wall Street, Room 5313, Ann Arbor, MI, 48105, USA
| | - Elif Arioglu Oral
- Brehm Center for Diabetes Research, Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan, 1000 Wall Street, Room 5313, Ann Arbor, MI, 48105, USA.
| |
Collapse
|
30
|
Zhou L, Yu M, Arshad M, Wang W, Lu Y, Gong J, Gu Y, Li P, Xu L. Coordination Among Lipid Droplets, Peroxisomes, and Mitochondria Regulates Energy Expenditure Through the CIDE-ATGL-PPARα Pathway in Adipocytes. Diabetes 2018; 67:1935-1948. [PMID: 29986925 DOI: 10.2337/db17-1452] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 06/29/2018] [Indexed: 11/13/2022]
Abstract
Metabolic homeostasis is maintained by an interplay among tissues, organs, intracellular organelles, and molecules. Cidea and Cidec are lipid droplet (LD)-associated proteins that promote lipid storage in brown adipose tissue (BAT) and white adipose tissue (WAT). Using ob/ob/Cidea-/- , ob/ob/Cidec-/- , and ob/ob/Cidea-/-/Cidec-/- mouse models and CIDE-deficient cells, we studied metabolic regulation during severe obesity to identify ways to maintain metabolic homeostasis and promote antiobesity effects. The phenotype of ob/ob/Cidea-/- mice was similar to that of ob/ob mice in terms of serum parameters, adipose tissues, lipid storage, and gene expression. Typical lipodystrophy accompanied by insulin resistance occurred in ob/ob/Cidec-/- mice, with ectopic storage of lipids in the BAT and liver. Interestingly, double deficiency of Cidea and Cidec activated both WAT and BAT to consume more energy and to increase insulin sensitivity compared with their behavior in the other three mouse models. Increased lipolysis, which occurred on the LD surfaces and released fatty acids, led to activated β-oxidation and oxidative phosphorylation in peroxisomes and mitochondria in CIDE-deficient adipocytes. The coordination among LDs, peroxisomes, and mitochondria was regulated by adipocyte triglyceride lipase (ATGL)-peroxisome proliferator-activated receptor α (PPARα). Double deficiency of Cidea and Cidec activated energy consumption in both WAT and BAT, which provided new insights into therapeutic approaches for obesity and diabetes.
Collapse
Affiliation(s)
- Linkang Zhou
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Miao Yu
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Muhammad Arshad
- Department of Bioinformatics and Biotechnology, International Islamic University, Islamabad, Pakistan
| | - Wenmin Wang
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Ye Lu
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Jingyi Gong
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Yangnan Gu
- Center for Plant Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Peng Li
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Li Xu
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, People's Republic of China
| |
Collapse
|
31
|
Godoy-Matos AF, Valerio CM, Moreira RO, Momesso DP, Bittencourt LK. Pancreatic fat deposition is increased and related to beta-cell function in women with familial partial lipodystrophy. Diabetol Metab Syndr 2018; 10:71. [PMID: 30275911 PMCID: PMC6158829 DOI: 10.1186/s13098-018-0375-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/24/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND To study pancreatic fat deposition and beta-cell function in familial partial lipodystrophy (FPLD) patients. METHODS In a cross-sectional study, eleven patients with FPLD, and eight healthy volunteers were matched for age and body mass index and studied at a referral center. Body composition was assessed using dual-energy X-ray absorptiometry and the Dixon method of magnetic resonance imaging was used to quantify pancreatic and liver fat. Fasting plasma glucose, insulin, leptin, lipids and homeostasis model assessment of insulin resistance values were measured, and an oral glucose tolerance test was performed. The insulinogenic index, Matsuda insulin sensitivity index and beta-cell disposition index were calculated. RESULTS The FPLD group presented a higher waist-to-hip ratio and fat mass ratio and lower total, truncal and lower-limb fat masses. Pancreatic and liver fat contents (log transformed) were significantly higher in the FPLD group (5.26 ± 1.5 vs. 4.08 ± 0.64, p = 0.034 and 0.77 ± 0.50 vs. 0.41 ± 0.18, p = 0.056, respectively). Pancreatic fat was inversely related to the DI (r = - 0.53, p = 0.027) and HDL-cholesterol (r = - 0.63, p = 0.003) and directly related to WHR (r = 0.60; p = 0.009), HbA1c (r = 0.58; p = 0.01) and serum triglyceride (r = 0.48, p = 0.034). Higher triglyceride and lower HDL-cholesterol levels were observed in the FPLD group. CONCLUSIONS This study demonstrated for the first time that pancreatic fat deposition is increased in FPLD. Moreover, an inverse relationship was demonstrated between pancreatic fat and beta-cell function. The findings of this study may be consistent with the expandability hypothesis and the twin cycle hypothesis.
Collapse
Affiliation(s)
- Amelio F. Godoy-Matos
- Serviço de Metabologia, Instituto Estadual de Diabetes e Endocrinologia (IEDE), Rua Visconde Silva, 52/1135 Botafogo, Rio de Janeiro, 22271-090 Brazil
| | - Cynthia M. Valerio
- Serviço de Metabologia, Instituto Estadual de Diabetes e Endocrinologia (IEDE), Rua Visconde Silva, 52/1135 Botafogo, Rio de Janeiro, 22271-090 Brazil
| | - Rodrigo O. Moreira
- Serviço de Metabologia, Instituto Estadual de Diabetes e Endocrinologia (IEDE), Rua Visconde Silva, 52/1135 Botafogo, Rio de Janeiro, 22271-090 Brazil
| | - Denise P. Momesso
- Serviço de Metabologia, Instituto Estadual de Diabetes e Endocrinologia (IEDE), Rua Visconde Silva, 52/1135 Botafogo, Rio de Janeiro, 22271-090 Brazil
| | - Leonardo K. Bittencourt
- Departamento de Radiologia, Universidade Federal Fluminense-Section Head, Abdominal and Pelvic Imaging-CDPI Clinics, DASA Company, Rio de Janeiro, Brazil
| |
Collapse
|
32
|
Affiliation(s)
- Saverio Cinti
- Professor of Human Anatomy, Director, Center of Obesity, University of Ancona (Politecnica delle Marche), Ancona, Italy
| |
Collapse
|
33
|
Sohn JH, Lee YK, Han JS, Jeon YG, Kim JI, Choe SS, Kim SJ, Yoo HJ, Kim JB. Perilipin 1 (Plin1) deficiency promotes inflammatory responses in lean adipose tissue through lipid dysregulation. J Biol Chem 2018; 293:13974-13988. [PMID: 30042231 DOI: 10.1074/jbc.ra118.003541] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/18/2018] [Indexed: 12/11/2022] Open
Abstract
Lipid droplets are specialized cellular organelles that contain neutral lipid metabolites and play dynamic roles in energy homeostasis. Perilipin 1 (Plin1), one of the major lipid droplet-binding proteins, is highly expressed in adipocytes. In mice, Plin1 deficiency impairs peripheral insulin sensitivity, accompanied with reduced fat mass. However, the mechanisms underlying insulin resistance in lean Plin1 knockout (Plin1-/-) mice are largely unknown. The current study demonstrates that Plin1 deficiency promotes inflammatory responses and lipolysis in adipose tissue, resulting in insulin resistance. M1-type adipose tissue macrophages (ATMs) were higher in Plin1-/- than in Plin1+/+ mice on normal chow diet. Moreover, using lipidomics analysis, we discovered that Plin1-/- adipocytes promoted secretion of pro-inflammatory lipid metabolites such as prostaglandins, which potentiated monocyte migration. In lean Plin1-/- mice, insulin resistance was relieved by macrophage depletion with clodronate, implying that elevated pro-inflammatory ATMs might be attributable for insulin resistance under Plin1 deficiency. Together, these data suggest that Plin1 is required to restrain fat loss and pro-inflammatory responses in adipose tissue by reducing futile lipolysis to maintain metabolic homeostasis.
Collapse
Affiliation(s)
- Jee Hyung Sohn
- From the National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul KS013 and
| | - Yun Kyung Lee
- From the National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul KS013 and
| | - Ji Seul Han
- From the National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul KS013 and
| | - Yong Geun Jeon
- From the National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul KS013 and
| | - Jong In Kim
- From the National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul KS013 and
| | - Sung Sik Choe
- From the National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul KS013 and
| | - Su Jung Kim
- the Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul KS013, South Korea
| | - Hyun Ju Yoo
- the Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul KS013, South Korea
| | - Jae Bum Kim
- From the National Creative Research Initiatives Center for Adipose Tissue Remodeling, Institute of Molecular Biology and Genetics, Department of Biological Sciences, Seoul National University, Seoul KS013 and
| |
Collapse
|
34
|
PPARγ Controls Ectopic Adipogenesis and Cross-Talks with Myogenesis During Skeletal Muscle Regeneration. Int J Mol Sci 2018; 19:ijms19072044. [PMID: 30011852 PMCID: PMC6073847 DOI: 10.3390/ijms19072044] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/06/2018] [Accepted: 07/09/2018] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscle is a regenerative tissue which can repair damaged myofibers through the activation of tissue-resident muscle stem cells (MuSCs). Many muscle diseases with impaired regeneration cause excessive adipose tissue accumulation in muscle, alter the myogenic fate of MuSCs, and deregulate the cross-talk between MuSCs and fibro/adipogenic progenitors (FAPs), a bi-potent cell population which supports myogenesis and controls intra-muscular fibrosis and adipocyte formation. In order to better characterize the interaction between adipogenesis and myogenesis, we studied muscle regeneration and MuSC function in whole body Pparg null mice generated by epiblast-specific Cre/lox deletion (PpargΔ/Δ). We demonstrate that deletion of PPARγ completely abolishes ectopic muscle adipogenesis during regeneration and impairs MuSC expansion and myogenesis after injury. Ex vivo assays revealed that perturbed myogenesis in PpargΔ/Δ mice does not primarily result from intrinsic defects of MuSCs or from perturbed myogenic support from FAPs. The immune transition from a pro- to anti-inflammatory MuSC niche during regeneration is perturbed in PpargΔ/Δ mice and suggests that PPARγ signaling in macrophages can interact with ectopic adipogenesis and influence muscle regeneration. Altogether, our study demonstrates that a PPARγ-dependent adipogenic response regulates muscle fat infiltration during regeneration and that PPARγ is required for MuSC function and efficient muscle repair.
Collapse
|
35
|
Girousse A, Virtue S, Hart D, Vidal-Puig A, Murgatroyd PR, Mouisel E, Sengenès C, Savage DB. Surplus fat rapidly increases fat oxidation and insulin resistance in lipodystrophic mice. Mol Metab 2018; 13:24-29. [PMID: 29789270 PMCID: PMC6026316 DOI: 10.1016/j.molmet.2018.05.006] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/02/2018] [Accepted: 05/07/2018] [Indexed: 11/24/2022] Open
Abstract
Objective Surplus dietary fat cannot be converted into other macronutrient forms or excreted, so has to be stored or oxidized. Healthy mammals store excess energy in the form of triacylgycerol (TAG) in lipid droplets within adipocytes rather than oxidizing it, and thus ultimately gain weight. The ‘overflow hypothesis’ posits that the capacity to increase the size and number of adipocytes is finite and that when this limit is exceeded, fat accumulates in ectopic sites and leads to metabolic disease. Methods Here we studied the energetic and biochemical consequences of short-term (2-day) excess fat ingestion in a lipodystrophic (A-ZIP/F-1) mouse model in which adipose capacity is severely restricted. Results In wildtype littermates, this acute exposure to high fat diets resulted in excess energy intake and weight gain without any significant changes in macronutrient oxidation rates, glucose, TAG, or insulin levels. In contrast, hyperphagic lipodystrophic mice failed to gain weight; rather, they significantly increased hepatic steatosis and fat oxidation. This response was associated with a significant increase in hyperglycemia, hyperinsulinemia, glucosuria, hypertriglyceridemia, and worsening insulin tolerance. Conclusions These data suggest that when adipose storage reserves are saturated, excess fat intake necessarily increases fat oxidation and induces oxidative substrate competition which exacerbates insulin resistance resolving any residual energy surplus through excretion of glucose. In contrast to wild type mice, lipodystrophic Azip mice manifest striking metabolic inflexibility. Wild type mice respond to excess dietary fat by storing the surplus energy and thus gain weight. In contrast, Azip mice increase fat oxidation and energy expenditure in response to surplus energy intake. Unfortunately this apparently ‘helpful’ adaptation also results in substrate competition and worsening insulin resistance.
Collapse
Affiliation(s)
- Amandine Girousse
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, Cambridge, CB2 0QQ, United Kingdom
| | - Samuel Virtue
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, Cambridge, CB2 0QQ, United Kingdom
| | - Dan Hart
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, Cambridge, CB2 0QQ, United Kingdom
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, Cambridge, CB2 0QQ, United Kingdom
| | - Peter R Murgatroyd
- NIHR/Wellcome Trust Clinical Research Facility, Addenbrooke's Hospital, Cambridge, UK
| | - Etienne Mouisel
- Institute of Cardiovascular and Metabolic Diseases, 31432, Toulouse, France
| | - Coralie Sengenès
- StromaLab, Paul Sabatier Toulouse University, ERL5311, 31432, Toulouse, France
| | - David B Savage
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, Cambridge, CB2 0QQ, United Kingdom.
| |
Collapse
|
36
|
Gallo-Ebert C, Francisco J, Liu HY, Draper R, Modi K, Hayward MD, Jones BK, Buiakova O, McDonough V, Nickels JT. Mice lacking ARV1 have reduced signs of metabolic syndrome and non-alcoholic fatty liver disease. J Biol Chem 2018; 293:5956-5974. [PMID: 29491146 DOI: 10.1074/jbc.ra117.000800] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 02/27/2018] [Indexed: 12/13/2022] Open
Abstract
Metabolic syndrome (MetS) is a term used to characterize individuals having at least three of the following diseases: obesity, dyslipidemia, hyperglycemia, insulin resistance, hypertension, and nonalcoholic fatty liver disease (NAFLD). It is widespread, and the number of individuals with MetS is increasing. However, the events leading to the manifestation of MetS are not well-understood. Here, we show that loss of murine ARV1 (mARV1) results in resistance to acquiring diseases associated with MetS. Arv1-/- animals fed a high-fat diet were resistant to diet-induced obesity, had lower blood cholesterol and triglyceride levels, and retained glucose tolerance and insulin sensitivity. Livers showed no gross morphological changes, contained lower levels of cholesterol, triglycerides, and fatty acids, and showed fewer signs of NAFLD. Knockout animals had elevated levels of liver farnesol X receptor (FXR) protein and its target, small heterodimer protein (SHP). They also had decreased levels of CYP7α1, CYP8β1, and mature SREBP1 protein, evidence suggesting that liver FXR signaling was activated. Strengthening this hypothesis was the fact that peroxisome proliferator-activating receptor α (PPARα) protein was elevated, along with its target, fibroblast growth factor 21 (FGF21). Arv1-/- animals excreted more fecal cholesterol, free fatty acids, and bile acids. Their small intestines had 1) changes in bile acid composition, 2) an increase in the level of the intestinal FXR antagonist, tauromuricholic acid, and 3) showed signs of attenuated FXR signaling. Overall, we believe that ARV1 function is deleterious when consuming a high-fat diet. We further hypothesize that ARV1 is critical for initiating events required for the progression of diseases associated with MetS and NAFLD.
Collapse
Affiliation(s)
- Christina Gallo-Ebert
- From the Institute of Metabolic Disorders, Genesis Biotechnology Group, Hamilton, New Jersey 08691
| | - Jamie Francisco
- From the Institute of Metabolic Disorders, Genesis Biotechnology Group, Hamilton, New Jersey 08691
| | - Hsing-Yin Liu
- From the Institute of Metabolic Disorders, Genesis Biotechnology Group, Hamilton, New Jersey 08691
| | | | - Kinnari Modi
- From the Institute of Metabolic Disorders, Genesis Biotechnology Group, Hamilton, New Jersey 08691
| | - Michael D Hayward
- Invivotek, Genesis Biotechnology Group, Hamilton, New Jersey 08691, and
| | - Beverly K Jones
- Invivotek, Genesis Biotechnology Group, Hamilton, New Jersey 08691, and
| | - Olesia Buiakova
- Invivotek, Genesis Biotechnology Group, Hamilton, New Jersey 08691, and
| | | | - Joseph T Nickels
- From the Institute of Metabolic Disorders, Genesis Biotechnology Group, Hamilton, New Jersey 08691, .,the Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, New Jersey 08901
| |
Collapse
|
37
|
Saja MF, Cook HT, Ruseva MM, Szajna M, Pickering MC, Woollard KJ, Botto M. A triglyceride-rich lipoprotein environment exacerbates renal injury in the accelerated nephrotoxic nephritis model. Clin Exp Immunol 2018; 192:337-347. [PMID: 29405270 PMCID: PMC5980512 DOI: 10.1111/cei.13111] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2018] [Indexed: 12/11/2022] Open
Abstract
Hyperlipidaemia accompanies chronic renal disease either as a consequence of the renal dysfunction or as part of generalized metabolic derangements. Under both situations, the lipid profile is characterized by accumulation of triglyceride-rich lipoproteins (TGRLs). This lipid profile is recognized as a risk factor for cardiovascular complications. Whether it may pose a risk for renal injury as well remains unclear. A hyper-TGRL state was generated in C57BL/6 mice using poloxamer-407 (P-407) and immune complex-mediated renal injury was triggered using the accelerated nephrotoxic nephritis (ANTN) model. The hyper-TGRL animals were hypersensitive to ANTN demonstrated by greater haematuria and glomerular cellularity. These changes were accompanied by increased glomerular accumulation of CD68+ macrophages. The hypersensitive response to ANTN was not seen in low-density lipoprotein receptor knock-out mice fed with a high fat diet, where triglyceride levels were lower but cholesterol levels comparable to those obtained using P-407. These data indicate that a hyper-TGRL state might be more detrimental to the kidneys than low-density lipoprotein-driven hypercholesterolaemia during immune complex-mediated nephritis. We speculate that the hyper-TGRL environment primes the kidney to exacerbated renal damage following an inflammatory insult with increased accumulation of macrophages that may play a key role in mediating the injurious effects.
Collapse
Affiliation(s)
- M F Saja
- Department of Medicine, Imperial College London, London, UK
| | - H T Cook
- Department of Medicine, Imperial College London, London, UK
| | - M M Ruseva
- Department of Medicine, Imperial College London, London, UK
| | - M Szajna
- Department of Medicine, Imperial College London, London, UK
| | - M C Pickering
- Department of Medicine, Imperial College London, London, UK
| | - K J Woollard
- Department of Medicine, Imperial College London, London, UK
| | - M Botto
- Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
38
|
Lee SY, Lee HY, Song JH, Kim GT, Jeon S, Song YJ, Lee JS, Hur JH, Oh HH, Park SY, Shim SM, Yoo HJ, Lee BC, Jiang XC, Choi CS, Park TS. Adipocyte-Specific Deficiency of De Novo Sphingolipid Biosynthesis Leads to Lipodystrophy and Insulin Resistance. Diabetes 2017; 66:2596-2609. [PMID: 28698261 PMCID: PMC7970771 DOI: 10.2337/db16-1232] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 06/28/2017] [Indexed: 12/16/2022]
Abstract
Sphingolipids have been implicated in the etiology of chronic metabolic diseases. Here, we investigated whether sphingolipid biosynthesis is associated with the development of adipose tissues and metabolic diseases. SPTLC2, a subunit of serine palmitoyltransferase, was transcriptionally upregulated in the adipose tissues of obese mice and in differentiating adipocytes. Adipocyte-specific SPTLC2-deficient (aSPTLC2 KO) mice had markedly reduced adipose tissue mass. Fatty acids that were destined for the adipose tissue were instead shunted to liver and caused hepatosteatosis. This impaired fat distribution caused systemic insulin resistance and hyperglycemia, indicating severe lipodystrophy. Mechanistically, sphingosine 1-phosphate (S1P) was reduced in the adipose tissues of aSPTLC2 KO mice, and this inhibited adipocyte proliferation and differentiation via the downregulation of S1P receptor 1 and decreased activity of the peroxisome proliferator-activator receptor γ. In addition, downregulation of SREBP (sterol regulatory element-binding protein)-1c prevented adipogenesis of aSPTLC2 KO adipocytes. Collectively, our observations suggest that the tight regulation of de novo sphingolipid biosynthesis and S1P signaling plays an important role in adipogenesis and hepatosteatosis.
Collapse
Affiliation(s)
- Su-Yeon Lee
- Department of Life Science, Gachon University, Sungnam, Korea
| | - Hui-Young Lee
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University School of Medicine, Incheon, Korea
| | - Jae-Hwi Song
- Department of Life Science, Gachon University, Sungnam, Korea
| | - Goon-Tae Kim
- Department of Life Science, Gachon University, Sungnam, Korea
| | - Suwon Jeon
- Department of Life Science, Gachon University, Sungnam, Korea
| | - Yoo-Jeong Song
- Department of Life Science, Gachon University, Sungnam, Korea
| | - Jae Sung Lee
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University School of Medicine, Incheon, Korea
| | - Jang-Ho Hur
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University School of Medicine, Incheon, Korea
| | - Hyun Hee Oh
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University School of Medicine, Incheon, Korea
| | - Shi-Young Park
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University School of Medicine, Incheon, Korea
| | - Soon-Mi Shim
- Department of Food Science and Technology, Sejong University, Seoul, Korea
| | - Hyun Joo Yoo
- Biomedical Research Center, Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Byung Cheon Lee
- College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Xian-Cheng Jiang
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, New York
| | - Cheol Soo Choi
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University School of Medicine, Incheon, Korea
- Endocrinology, Internal Medicine, Gachon University Gil Medical Center, Incheon, Korea
| | - Tae-Sik Park
- Department of Life Science, Gachon University, Sungnam, Korea
| |
Collapse
|
39
|
Kimmel AR, Sztalryd C. The Perilipins: Major Cytosolic Lipid Droplet-Associated Proteins and Their Roles in Cellular Lipid Storage, Mobilization, and Systemic Homeostasis. Annu Rev Nutr 2017; 36:471-509. [PMID: 27431369 DOI: 10.1146/annurev-nutr-071813-105410] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The discovery by Dr. Constantine Londos of perilipin 1, the major scaffold protein at the surface of cytosolic lipid droplets in adipocytes, marked a fundamental conceptual change in the understanding of lipolytic regulation. Focus then shifted from the enzymatic activation of lipases to substrate accessibility, mediated by perilipin-dependent protein sequestration and recruitment. Consequently, the lipid droplet became recognized as a unique, metabolically active cellular organelle and its surface as the active site for novel protein-protein interactions. A new area of investigation emerged, centered on lipid droplets' biology and their role in energy homeostasis. The perilipin family is of ancient origin and has expanded to include five mammalian genes and a growing list of evolutionarily conserved members. Universally, the perilipins modulate cellular lipid storage. This review provides a summary that connects the perilipins to both cellular and whole-body homeostasis.
Collapse
Affiliation(s)
- Alan R Kimmel
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, The National Institutes of Health, Bethesda, Maryland 20892;
| | - Carole Sztalryd
- The Geriatric Research Education and Clinical Center, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland 21201.,Division of Endocrinology, Department of Medicine, School of Medicine, University of Maryland, Baltimore, Maryland 21201;
| |
Collapse
|
40
|
Rozo AV, Babu DA, Suen PA, Groff DN, Seeley RJ, Simmons RA, Seale P, Ahima RS, Stoffers DA. Neonatal GLP1R activation limits adult adiposity by durably altering hypothalamic architecture. Mol Metab 2017; 6:748-759. [PMID: 28702330 PMCID: PMC5485307 DOI: 10.1016/j.molmet.2017.05.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/03/2017] [Accepted: 05/10/2017] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Adult obesity risk is influenced by alterations to fetal and neonatal environments. Modifying neonatal gut or neurohormone signaling pathways can have negative metabolic consequences in adulthood. Here we characterize the effect of neonatal activation of glucagon like peptide-1 (GLP-1) receptor (GLP1R) signaling on adult adiposity and metabolism. METHODS Wild type C57BL/6 mice were injected with 1 nmol/kg Exendin-4 (Ex-4), a GLP1R agonist, for 6 consecutive days after birth. Growth, body composition, serum analysis, energy expenditure, food intake, and brain and fat pad histology and gene expression were assessed at multiple time points through 42 weeks. Similar analyses were conducted in a Glp1r conditional allele crossed with a Sim1Cre deleter strain to produce Sim1Cre;Glp1rloxP/loxP mice and control littermates. RESULTS Neonatal administration of Ex-4 reduced adult body weight and fat mass, increased energy expenditure, and conferred protection from diet-induced obesity in female mice. This was associated with induction of brown adipose genes and increased noradrenergic fiber density in parametrial white adipose tissue (WAT). We further observed durable alterations in orexigenic and anorexigenic projections to the paraventricular hypothalamic nucleus (PVH). Genetic deletion of Glp1r in the PVH by Sim1-Cre abrogated the impact of neonatal Ex-4 on adult body weight, WAT browning, and hypothalamic architecture. CONCLUSION These observations suggest that the acute activation of GLP1R in neonates durably alters hypothalamic architecture to limit adult weight gain and adiposity, identifying GLP1R as a therapeutic target for obesity prevention.
Collapse
Affiliation(s)
- Andrea V. Rozo
- Institute for Diabetes, Obesity and Metabolism and the Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Daniella A. Babu
- Institute for Diabetes, Obesity and Metabolism and the Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - PoMan A. Suen
- Institute for Diabetes, Obesity and Metabolism and the Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - David N. Groff
- Institute for Diabetes, Obesity and Metabolism and the Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Randy J. Seeley
- Department of Surgery, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Rebecca A. Simmons
- Department of Pediatrics, Division of Neonatology, The Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Patrick Seale
- Institute for Diabetes, Obesity and Metabolism and the Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Rexford S. Ahima
- Institute for Diabetes, Obesity and Metabolism and the Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| | - Doris A. Stoffers
- Institute for Diabetes, Obesity and Metabolism and the Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| |
Collapse
|
41
|
Abulizi A, Perry RJ, Camporez JPG, Jurczak MJ, Petersen KF, Aspichueta P, Shulman GI. A controlled-release mitochondrial protonophore reverses hypertriglyceridemia, nonalcoholic steatohepatitis, and diabetes in lipodystrophic mice. FASEB J 2017; 31:2916-2924. [PMID: 28330852 DOI: 10.1096/fj.201700001r] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 03/06/2017] [Indexed: 12/31/2022]
Abstract
Lipodystrophy is a rare disorder characterized by complete or partial loss of adipose tissue. Patients with lipodystrophy exhibit hypertriglyceridemia, severe insulin resistance, type 2 diabetes, and nonalcoholic steatohepatitis (NASH). Efforts to ameliorate NASH in lipodystrophies with pharmacologic agents have met with limited success. We examined whether a controlled-release mitochondrial protonophore (CRMP) that produces mild liver-targeted mitochondrial uncoupling could decrease hypertriglyceridemia and reverse NASH and diabetes in a mouse model (fatless AZIP/F-1 mice) of severe lipodystrophy and diabetes. After 4 wk of oral CRMP (2 mg/kg body weight per day) or vehicle treatment, mice underwent hyperinsulinemic-euglycemic clamps combined with radiolabeled glucose to assess liver and muscle insulin responsiveness and tissue lipid measurements. CRMP treatment reversed hypertriglyceridemia and insulin resistance in liver and skeletal muscle. Reversal of insulin resistance could be attributed to reductions in diacylglycerol content and reduced PKC-ε and PKC-θ activity in liver and muscle respectively. CRMP treatment also reversed NASH as reflected by reductions in plasma aspartate aminotransferase and alanine aminotransferase concentrations; hepatic steatosis; and hepatic expression of IL-1α, -β, -2, -4, -6, -10, -12, CD69, and caspase 3 and attenuated activation of the IRE-1α branch of the unfolded protein response. Taken together, these results provide proof of concept for the development of liver-targeted mitochondrial uncoupling agents as a potential novel therapy for lipodystrophy-associated hypertriglyceridemia, NASH and diabetes.-Abulizi, A., Perry, R. J., Camporez, J. P. G., Jurczak, M. J., Petersen, K. F., Aspichueta, P., Shulman, G. I. A controlled-release mitochondrial protonophore reverses hypertriglyceridemia, nonalcoholic steatohepatitis, and diabetes in lipodystrophic mice.
Collapse
Affiliation(s)
- Abudukadier Abulizi
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Rachel J Perry
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - João Paulo G Camporez
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Michael J Jurczak
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kitt Falk Petersen
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Patricia Aspichueta
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Physiology, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), Leioa, Spain.,Biocruces Research Institute, Barakaldo, Spain
| | - Gerald I Shulman
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA; .,Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA.,Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
42
|
Langhi C, Arias N, Rajamoorthi A, Basta J, Lee RG, Baldán Á. Therapeutic silencing of fat-specific protein 27 improves glycemic control in mouse models of obesity and insulin resistance. J Lipid Res 2016; 58:81-91. [PMID: 27884961 PMCID: PMC5234712 DOI: 10.1194/jlr.m069799] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 11/16/2016] [Indexed: 12/21/2022] Open
Abstract
Obesity is a component of the metabolic syndrome, mechanistically linked to diabetes, fatty liver disease, and cardiovascular disease. Proteins that regulate the metabolic fate of intracellular lipid droplets are potential therapeutic candidates to treat obesity and its related consequences. CIDEC (cell death-inducing DFFA-like effector C), also known in mice as Fsp27 (fat-specific protein 27), is a lipid droplet-associated protein that prevents lipid mobilization and promotes intracellular lipid storage. The consequences of complete loss of FSP27 on hepatic metabolism and on insulin resistance are controversial, as both healthy and deleterious lipodystrophic phenotypes have been reported in Fsp27−/− mice. To test whether therapeutic silencing of Fsp27 might be useful to improve obesity, fatty liver, and glycemic control, we used antisense oligonucleotides (ASOs) in both nutritional (high-fat diet) and genetic (leptin-deficient ob/ob) mouse models of obesity, hyperglycemia, and hepatosteatosis. We show that partial silencing Fsp27 in either model results in the robust decrease in visceral fat, improved insulin sensitivity and whole-body glycemic control, and tissue-specific changes in transcripts controlling lipid oxidation and synthesis. These data suggest that partial reduction of FSP27 activity (e.g., using ASOs) might be exploited therapeutically in insulin-resistant obese or overweight patients.
Collapse
Affiliation(s)
- Cédric Langhi
- Edward A. Doisy Department of Biochemistry & Molecular Biology Saint Louis University, Saint Louis, MO 63104
| | - Noemí Arias
- Edward A. Doisy Department of Biochemistry & Molecular Biology Saint Louis University, Saint Louis, MO 63104
| | - Ananthi Rajamoorthi
- Edward A. Doisy Department of Biochemistry & Molecular Biology Saint Louis University, Saint Louis, MO 63104
| | - Jeannine Basta
- Department of Internal Medicine, Saint Louis University, Saint Louis, MO 63104
| | - Richard G Lee
- Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Carlsbad, CA 92010
| | - Ángel Baldán
- Edward A. Doisy Department of Biochemistry & Molecular Biology Saint Louis University, Saint Louis, MO 63104 .,Center for Cardiovascular Research Saint Louis University, Saint Louis, MO 63104.,Liver Center, Saint Louis University, Saint Louis, MO 63104
| |
Collapse
|
43
|
Wu CH, Mohammadmoradi S, Thompson J, Su W, Gong M, Nguyen G, Yiannikouris F. Adipocyte (Pro)Renin-Receptor Deficiency Induces Lipodystrophy, Liver Steatosis and Increases Blood Pressure in Male Mice. Hypertension 2016; 68:213-9. [PMID: 27185751 DOI: 10.1161/hypertensionaha.115.06954] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 04/18/2016] [Indexed: 01/13/2023]
Abstract
Adipose tissue dysfunction related to obesity is overwhelmingly associated with increased risk of developing cardiovascular diseases. In the setting of obesity, (pro)renin receptor (PRR) is increased in adipose tissue of mice. We sought to determine the physiological consequences of adipocyte-PRR deficiency using adiponectin-Cre mice. We report a unique model of adipocyte-PRR-deficient mice (PRR(Adi/Y)) with almost no detectable white adipose tissues. As a consequence, the livers of PRR(Adi/Y) mice were enlarged and demonstrated a marked accumulation of lipids. Adipocyte-specific deficiency of PRR increased systolic blood pressure and the concentration of soluble PRR in plasma. To determine whether adipocyte-PRR was involved in the development of obesity-induced hypertension, mice were fed a low-fat or a high-fat diet for 16 weeks. Adipocyte-PRR-deficient mice were resistant to diet-induced obesity. Both high-fat- and low-fat-fed PRR(Adi/Y) mice had elevated insulin levels. Interestingly, adipocyte-PRR deficiency improved glucose tolerance in high-fat-fed PRR(Adi/Y) mice. In response to feeding either low-fat or high-fat diets, systolic blood pressure was greater in PRR(Adi/Y) mice than in control mice. High-fat feeding elevated soluble PRR concentration in control and PRR(Adi/Y) mice. In vitro knockdown of PRR by siRNA significantly decreased mRNA abundance of PPARγ (peroxisome proliferator-activated receptor gamma), suggesting an important role for PRR in adipogenesis. Our data indicate that adipocyte-PRR is involved in lipid homeostasis and glucose and insulin homeostasis, and that soluble PRR may be a predictor of metabolic disturbances and play a role in systolic blood pressure regulation.
Collapse
Affiliation(s)
- Chia-Hua Wu
- From the Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., F.Y.), Division of Endocrinology and Molecular Medicine (J.T.), and Department of Physiology (W.S., M.G.), University of Kentucky, Lexington; and Institut National de la Santè et de la Recherche Mèdicale (INSERM) U489 and Collège de France, Experimental Medicine Unit, Paris, France (G.N.)
| | - Shayan Mohammadmoradi
- From the Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., F.Y.), Division of Endocrinology and Molecular Medicine (J.T.), and Department of Physiology (W.S., M.G.), University of Kentucky, Lexington; and Institut National de la Santè et de la Recherche Mèdicale (INSERM) U489 and Collège de France, Experimental Medicine Unit, Paris, France (G.N.)
| | - Joel Thompson
- From the Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., F.Y.), Division of Endocrinology and Molecular Medicine (J.T.), and Department of Physiology (W.S., M.G.), University of Kentucky, Lexington; and Institut National de la Santè et de la Recherche Mèdicale (INSERM) U489 and Collège de France, Experimental Medicine Unit, Paris, France (G.N.)
| | - Wen Su
- From the Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., F.Y.), Division of Endocrinology and Molecular Medicine (J.T.), and Department of Physiology (W.S., M.G.), University of Kentucky, Lexington; and Institut National de la Santè et de la Recherche Mèdicale (INSERM) U489 and Collège de France, Experimental Medicine Unit, Paris, France (G.N.)
| | - Ming Gong
- From the Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., F.Y.), Division of Endocrinology and Molecular Medicine (J.T.), and Department of Physiology (W.S., M.G.), University of Kentucky, Lexington; and Institut National de la Santè et de la Recherche Mèdicale (INSERM) U489 and Collège de France, Experimental Medicine Unit, Paris, France (G.N.)
| | - Genevieve Nguyen
- From the Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., F.Y.), Division of Endocrinology and Molecular Medicine (J.T.), and Department of Physiology (W.S., M.G.), University of Kentucky, Lexington; and Institut National de la Santè et de la Recherche Mèdicale (INSERM) U489 and Collège de France, Experimental Medicine Unit, Paris, France (G.N.)
| | - Frédérique Yiannikouris
- From the Department of Pharmacology and Nutritional Sciences (C.-H.W., S.M., F.Y.), Division of Endocrinology and Molecular Medicine (J.T.), and Department of Physiology (W.S., M.G.), University of Kentucky, Lexington; and Institut National de la Santè et de la Recherche Mèdicale (INSERM) U489 and Collège de France, Experimental Medicine Unit, Paris, France (G.N.).
| |
Collapse
|
44
|
Qiang G, Whang Kong H, Xu S, Pham HA, Parlee SD, Burr AA, Gil V, Pang J, Hughes A, Gu X, Fantuzzi G, MacDougald OA, Liew CW. Lipodystrophy and severe metabolic dysfunction in mice with adipose tissue-specific insulin receptor ablation. Mol Metab 2016; 5:480-490. [PMID: 27408774 PMCID: PMC4921803 DOI: 10.1016/j.molmet.2016.05.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 04/26/2016] [Accepted: 05/06/2016] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVE Insulin signaling plays pivotal roles in the development and metabolism of many tissues and cell types. A previous study demonstrated that ablation of insulin receptor (IR) with aP2-Cre markedly reduced adipose tissues mass and protected mice from obesity. However, multiple studies have demonstrated widespread non-adipocyte recombination of floxed alleles in aP2-Cre mice. These findings underscore the need to re-evaluate the role of IR in adipocyte and systemic metabolism with a more adipose tissue-specific Cre mouse line. METHODS We generated and phenotyped a new adipose tissue-specific IR mouse model using the adipose tissue-specific Adipoq-Cre line. RESULTS Here we show that the Adipoq-Cre-mediated IR KO in mice leads to lipodystrophy and metabolic dysfunction, which is in stark contrast to the previous study. In contrast to white adipocytes, absence of insulin signaling does not affect development of marrow and brown adipocytes, but instead is required for lipid accumulation particularly for the marrow adipocytes. Lipodystrophic IR KO mice have profound insulin resistance, hyperglycemia, organomegaly, and impaired adipokine secretion. CONCLUSIONS Our results demonstrate differential roles for insulin signaling for white, brown, and marrow adipocyte development and metabolic regulation.
Collapse
Affiliation(s)
- Guifen Qiang
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Hyerim Whang Kong
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Shanshan Xu
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Hoai An Pham
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Sebastian D Parlee
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Aaron A Burr
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Victoria Gil
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Jingbo Pang
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Amy Hughes
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Xuejiang Gu
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Giamila Fantuzzi
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Ormond A MacDougald
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Chong Wee Liew
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
45
|
Heterogeneity of white adipose tissue: molecular basis and clinical implications. Exp Mol Med 2016; 48:e215. [PMID: 26964831 PMCID: PMC4892883 DOI: 10.1038/emm.2016.5] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 11/29/2015] [Indexed: 02/08/2023] Open
Abstract
Adipose tissue is a highly heterogeneous endocrine organ. The heterogeneity among different anatomical depots stems from their intrinsic differences in cellular and physiological properties, including developmental origin, adipogenic and proliferative capacity, glucose and lipid metabolism, insulin sensitivity, hormonal control, thermogenic ability and vascularization. Additional factors that influence adipose tissue heterogeneity are genetic predisposition, environment, gender and age. Under obese condition, these depot-specific differences translate into specific fat distribution patterns, which are closely associated with differential cardiometabolic risks. For instance, individuals with central obesity are more susceptible to developing diabetes and cardiovascular complications, whereas those with peripheral obesity are more metabolically healthy. This review summarizes the clinical and mechanistic evidence for the depot-specific differences that give rise to different metabolic consequences, and provides therapeutic insights for targeted treatment of obesity.
Collapse
|
46
|
Huang-Doran I, Franks S. Genetic Rodent Models of Obesity-Associated Ovarian Dysfunction and Subfertility: Insights into Polycystic Ovary Syndrome. Front Endocrinol (Lausanne) 2016; 7:53. [PMID: 27375552 PMCID: PMC4894870 DOI: 10.3389/fendo.2016.00053] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/17/2016] [Indexed: 01/26/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrinopathy affecting women and a leading cause of female infertility worldwide. Defined clinically by the presence of hyperandrogenemia and oligomenorrhoea, PCOS represents a state of hormonal dysregulation, disrupted ovarian follicle dynamics, and subsequent oligo- or anovulation. The syndrome's prevalence is attributed, at least partly, to a well-established association with obesity and insulin resistance (IR). Indeed, the presence of severe PCOS in human genetic obesity and IR syndromes supports a causal role for IR in the pathogenesis of PCOS. However, the molecular mechanisms underlying this causality, as well as the important role of hyperandrogenemia, remain poorly elucidated. As such, treatment of PCOS is necessarily empirical, focusing on symptom alleviation. The generation of knockout and transgenic rodent models of obesity and IR offers a promising platform in which to address mechanistic questions about reproductive dysfunction in the context of metabolic disease. Similarly, the impact of primary perturbations in rodent gonadotrophin or androgen signaling has been interrogated. However, the insights gained from such models have been limited by the relatively poor fidelity of rodent models to human PCOS. In this mini review, we evaluate the ovarian phenotypes associated with rodent models of obesity and IR, including the extent of endocrine disturbance, ovarian dysmorphology, and subfertility. We compare them to both human PCOS and other animal models of the syndrome (genetic and hormonal), explore reasons for their discordance, and consider the new opportunities that are emerging to better understand and treat this important condition.
Collapse
Affiliation(s)
- Isabel Huang-Doran
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
- *Correspondence: Isabel Huang-Doran,
| | - Stephen Franks
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
47
|
Mann JP, Semple RK, Armstrong MJ. How Useful Are Monogenic Rodent Models for the Study of Human Non-Alcoholic Fatty Liver Disease? Front Endocrinol (Lausanne) 2016; 7:145. [PMID: 27899914 PMCID: PMC5110950 DOI: 10.3389/fendo.2016.00145] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 11/01/2016] [Indexed: 12/22/2022] Open
Abstract
Improving understanding of the genetic basis of human non-alcoholic fatty liver disease (NAFLD) has the potential to facilitate risk stratification of affected patients, permit personalized treatment, and inform development of new therapeutic strategies. Animal models have been widely used to interrogate the pathophysiology of, and genetic predisposition to, NAFLD. Nevertheless, considerable interspecies differences in intermediary metabolism potentially limit the extent to which results can be extrapolated to humans. For example, human genome-wide association studies have identified polymorphisms in PNPLA3 and TM6SF2 as the two most prevalent determinants of susceptibility to NAFLD and its inflammatory component (NASH), but animal models of these mutations have had only variable success in recapitulating this link. In this review, we critically appraise selected murine monogenic models of NAFLD, NASH, and hepatocellular carcinoma (HCC) with a focus on how closely they mirror human disease.
Collapse
Affiliation(s)
- Jake P. Mann
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Robert K. Semple
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK
- The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
- *Correspondence: Robert K. Semple,
| | - Matthew J. Armstrong
- Centre for Liver Research, National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, University of Birmingham, Birmingham, UK
- Liver Unit, Queen Elizabeth University Hospital Birmingham, Birmingham, UK
| |
Collapse
|
48
|
Grundy SM. Adipose tissue and metabolic syndrome: too much, too little or neither. Eur J Clin Invest 2015; 45:1209-17. [PMID: 26291691 PMCID: PMC5049481 DOI: 10.1111/eci.12519] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 08/15/2015] [Indexed: 12/17/2022]
Abstract
Obesity is strongly associated with metabolic syndrome. Recent research suggests that excess adipose tissue plays an important role in development of the syndrome. On the other hand, persons with a deficiency of adipose tissue (e.g. lipodystrophy) also manifest the metabolic syndrome. In some animal models, expansion of adipose tissue pools mitigates adverse metabolic components (e.g. insulin resistance, hyperglycaemia and dyslipidemia). Hence, there are conflicting data as to whether adipose tissue worsens the metabolic syndrome or protects against it. This conflict may relate partly to locations of adipose tissue pools. For instance, lower body adipose tissue may be protective whereas upper body adipose tissue may promote the syndrome. One view holds that in either case, the accumulation of ectopic fat in muscle and liver is the driving factor underlying the syndrome. If so, there may be some link between adipose tissue fat and ectopic fat. But the mechanisms underlying this connection are not clear. A stronger association appears to exist between excessive caloric intake and ectopic fat accumulation. Adipose tissue may act as a buffer to reduce the impact of excess energy consumption by fat storage; but once a constant weight has been achieved, it is unclear whether adipose tissue influences levels of ectopic fat. Another mechanism whereby adipose tissue could worsen the metabolic syndrome is through release of adipokines. This is an intriguing mechanism, but the impact of adipokines on metabolic syndrome risk factors is uncertain. Thus, many potential connections between adipose tissue and metabolic syndrome remain to unravelled.
Collapse
Affiliation(s)
- Scott M Grundy
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Veterans Affairs Medical Center, Dallas, TX, USA
| |
Collapse
|
49
|
HMGA1 overexpression in adipose tissue impairs adipogenesis and prevents diet-induced obesity and insulin resistance. Sci Rep 2015; 5:14487. [PMID: 26411793 PMCID: PMC4585969 DOI: 10.1038/srep14487] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 08/12/2015] [Indexed: 01/14/2023] Open
Abstract
High-Mobility-Group-A1 (HMGA1) proteins are non-histone proteins that regulate chromatin structure and gene expression during embryogenesis, tumourigenesis and immune responses. In vitro studies suggest that HMGA1 proteins may be required to regulate adipogenesis. To examine the role of HMGA1 in vivo, we generated transgenic mice overexpressing HMGA1 in adipose tissues. HMGA1 transgenic mice showed a marked reduction in white and brown adipose tissue mass that was associated with downregulation of genes involved in adipogenesis and concomitant upregulation of preadipocyte markers. Reduced adipogenesis and decreased fat mass were not associated with altered glucose homeostasis since HMGA1 transgenic mice fed a regular-chow diet exhibited normal glucose tolerance and insulin sensitivity. However, when fed a high-fat diet, overexpression of HMGA1 resulted in decreased body-weight gain, reduced fat mass, but improved insulin sensitivity and glucose tolerance. Although HMGA1 transgenic mice exhibited impaired glucose uptake in adipose tissue due to impaired adipogenesis, the increased glucose uptake observed in skeletal muscle may account for the improved glucose homeostasis. Our results indicate that HMGA1 plays an important function in the regulation of white and brown adipogenesis in vivo and suggests that impaired adipocyte differentiation and decreased fat mass is not always associated with impaired whole-body glucose homeostasis.
Collapse
|
50
|
Akinci B, Koseoglu FD, Onay H, Yavuz S, Altay C, Simsir IY, Ozisik S, Demir L, Korkut M, Yilmaz N, Ozen S, Akinci G, Atik T, Calan M, Secil M, Comlekci A, Demir T. Acquired partial lipodystrophy is associated with increased risk for developing metabolic abnormalities. Metabolism 2015; 64:1086-95. [PMID: 26139569 DOI: 10.1016/j.metabol.2015.06.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 06/01/2015] [Accepted: 06/04/2015] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Acquired partial lipodystrophy (APL) is a rare disorder characterized by progressive selective fat loss. In previous studies, metabolic abnormalities were reported to be relatively rare in APL, whilst they were quite common in other types of lipodystrophy syndromes. METHODS In this nationwide cohort study, we evaluated 21 Turkish patients with APL who were enrolled in a prospective follow-up protocol. Subjects were investigated for metabolic abnormalities. Fat distribution was assessed by whole body MRI. Hepatic steatosis was evaluated by ultrasound, MRI and MR spectroscopy. Patients with diabetes underwent a mix meal stimulated C-peptide/insulin test to investigate pancreatic beta cell functions. Leptin and adiponectin levels were measured. RESULTS Fifteen individuals (71.4%) had at least one metabolic abnormality. Six patients (28.6%) had diabetes, 12 (57.1%) hypertrigylceridemia, 10 (47.6%) low HDL cholesterol, and 11 (52.4%) hepatic steatosis. Steatohepatitis was further confirmed in 2 patients with liver biopsy. Anti-GAD was negative in all APL patients with diabetes. APL patients with diabetes had lower leptin and adiponectin levels compared to patients with type 2 diabetes and healthy controls. However, contrary to what we observed in patients with congenital generalized lipodystrophy (CGL), we did not detect consistently very low leptin levels in APL patients. The mix meal test suggested that APL patients with diabetes had a significant amount of functional pancreatic beta cells, and their diabetes was apparently associated with insulin resistance. CONCLUSIONS Our results show that APL is associated with increased risk for developing metabolic abnormalities. We suggest that close long-term follow-up is required to identify and manage metabolic abnormalities in APL.
Collapse
Affiliation(s)
- Baris Akinci
- Dokuz Eylul University, Division of Endocrinology, Izmir, Turkey.
| | | | - Huseyin Onay
- Ege University, Department of Medical Genetics, Izmir, Turkey
| | - Sevgi Yavuz
- Kanuni Sultan Suleyman Training Hospital, Department of Dermatology, Istanbul, Turkey
| | - Canan Altay
- Dokuz Eylul University, Department of Radiology, Izmir, Turkey
| | | | - Secil Ozisik
- Dokuz Eylul University, Division of Endocrinology, Izmir, Turkey
| | - Leyla Demir
- Ataturk Training Hospital, Department of Biochemistry, Izmir, Turkey
| | - Meltem Korkut
- Yeditepe University, Division of Pediatric Gastroenterology, Istanbul, Turkey
| | - Nusret Yilmaz
- Akdeniz University, Division of Endocrinology, Antalya, Turkey
| | - Samim Ozen
- Ege University, Department of Medical Genetics, Izmir, Turkey; Ege University, Division of Pediatric Endocrinology, Izmir, Turkey
| | - Gulcin Akinci
- Dr.Behcet Uz Children's Hospital, Division of Pediatric Neurology, Izmir, Turkey
| | - Tahir Atik
- Ege University, Department of Medical Genetics, Izmir, Turkey
| | - Mehmet Calan
- Dokuz Eylul University, Division of Endocrinology, Izmir, Turkey
| | - Mustafa Secil
- Dokuz Eylul University, Department of Radiology, Izmir, Turkey
| | | | - Tevfik Demir
- Dokuz Eylul University, Division of Endocrinology, Izmir, Turkey
| |
Collapse
|