1
|
Wang Y, Hong S, Hudson H, Kory N, Kinch LN, Kozlitina J, Cohen JC, Hobbs HH. PNPLA3(148M) is a gain-of-function mutation that promotes hepatic steatosis by inhibiting ATGL-mediated triglyceride hydrolysis. J Hepatol 2025; 82:871-881. [PMID: 39550037 DOI: 10.1016/j.jhep.2024.10.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND & AIMS PNPLA3(148M) (patatin-like phospholipase domain-containing protein 3) is the most impactful genetic risk factor for steatotic liver disease. A key unresolved issue is whether PNPLA3(148M) confers a loss- or gain-of-function. Here we test the hypothesis that PNPLA3 causes steatosis by sequestering ABHD5 (α/β hydrolase domain-containing protein 5), the cofactor of ATGL (adipose TG lipase), thus limiting mobilization of hepatic triglyceride (TG). METHODS We quantified and compared the physical interactions between ABHD5 and PNPLA3/ATGL in cultured hepatocytes using NanoBiT complementation assays and immunocytochemistry. Recombinant proteins purified from human cells were used to compare TG hydrolytic activities of PNPLA3 and ATGL in the presence or absence of ABHD5. Adenoviruses and adeno-associated viruses were used to express PNPLA3 in liver-specific Atgl-/- mice and to express ABHD5 in livers of Pnpla3M/M mice, respectively. RESULTS ABHD5 interacted preferentially with PNPLA3 relative to ATGL in cultured hepatocytes. No differences were seen in the strength of the interactions between ABHD5 with PNPLA3(WT) and PNPLA3(148M). In contrast to prior findings, we found that PNPLA3, like ATGL, is activated by ABHD5 in in vitro assays using purified proteins. PNPLA3(148M)-associated inhibition of TG hydrolysis required that ATGL be expressed and that PNPLA3 be located on lipid droplets. Finally, overexpression of ABHD5 reversed the hepatic steatosis in Pnpla3M/M mice. CONCLUSIONS These findings support the premise that PNPLA3(148M) is a gain-of-function mutation that promotes hepatic steatosis by accumulating on lipid droplets and inhibiting ATGL-mediated lipolysis in an ABHD5-dependent manner. Our results predict that reducing, rather than increasing, PNPLA3 expression will be the best strategy to treat PNPLA3(148M)-associated steatotic liver disease. IMPACT AND IMPLICATIONS Steatotic liver disease (SLD) is a common complex disorder associated with both environmental and genetic risk factors. PNPLA3(148M) is the most impactful genetic risk factor for SLD and yet its pathogenic mechanism remains controversial. Herein, we provide evidence that PNPLA3(148M) promotes triglyceride (TG) accumulation by sequestering ABHD5, thus limiting its availability to activate ATGL. Although the substitution of methionine for isoleucine reduces the TG hydrolase activity of PNPLA3, the loss of enzymatic function is not directly related to the steatotic effect of the variant. It is the resulting accumulation of PNPLA3 on LDs that confers a gain-of-function by interfering with ATGL-mediated TG hydrolysis. These findings have implications for the design of potential PNPLA3(148M)-based therapies. Reducing, rather than increasing, PNPLA3 levels is predicted to reverse steatosis in susceptible individuals.
Collapse
Affiliation(s)
- Yang Wang
- Department of Molecular Genetics, University of Texas Southwestern Medical Center (UTSW), Dallas, TX 75390-9046, USA.
| | - Sen Hong
- Department of Molecular Genetics, University of Texas Southwestern Medical Center (UTSW), Dallas, TX 75390-9046, USA; Howard Hughes Medical Institute, UTSW, Dallas, TX 75390, USA
| | - Hannah Hudson
- Department of Molecular Genetics, University of Texas Southwestern Medical Center (UTSW), Dallas, TX 75390-9046, USA
| | - Nora Kory
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Lisa N Kinch
- Department of Molecular Genetics, University of Texas Southwestern Medical Center (UTSW), Dallas, TX 75390-9046, USA; Howard Hughes Medical Institute, UTSW, Dallas, TX 75390, USA
| | - Julia Kozlitina
- The Eugene McDermott Center for Human Growth and Development, UTSW, Dallas, TX, 75390, USA
| | - Jonathan C Cohen
- Department of Molecular Genetics, University of Texas Southwestern Medical Center (UTSW), Dallas, TX 75390-9046, USA; Center for Human Nutrition, UTSW, Dallas, TX 75390, USA
| | - Helen H Hobbs
- Department of Molecular Genetics, University of Texas Southwestern Medical Center (UTSW), Dallas, TX 75390-9046, USA; Howard Hughes Medical Institute, UTSW, Dallas, TX 75390, USA; The Eugene McDermott Center for Human Growth and Development, UTSW, Dallas, TX, 75390, USA.
| |
Collapse
|
2
|
Mouisel E, Bodon A, Noll C, Cassant-Sourdy S, Marques MA, Flores-Flores R, Riant E, Bergoglio C, Vezin P, Caspar-Bauguil S, Fournes-Fraresso C, Tavernier G, Oumar KAI, Gourdy P, Blondin DP, Denechaud PD, Carpentier AC, Langin D. Cold-induced thermogenesis requires neutral-lipase-mediated intracellular lipolysis in brown adipocytes. Cell Metab 2025; 37:429-440.e5. [PMID: 39566492 DOI: 10.1016/j.cmet.2024.10.018] [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: 02/02/2024] [Revised: 07/01/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024]
Abstract
Long-chain fatty acids (FAs) are the major substrates fueling brown adipose tissue (BAT) thermogenesis. Investigation of mouse models has previously called into question the contribution of brown adipocyte intracellular lipolysis to cold-induced non-shivering thermogenesis. Here, we determined the role of the lipolytic enzymes, adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL), in BAT thermogenesis. Brown fat from mice with inducible brown-adipocyte-specific deletion of ATGL and HSL (BAHKO) is hypertrophied with increased lipid droplet size and preserved mitochondria area and density. Maintenance of body temperature during cold exposure is compromised in BAHKO mice in the fasted but not in the fed state. This altered response to cold is observed in various thermal and nutritional conditions. Positron emission tomography-computed tomography using [11C]-acetate and [11C]-palmitate shows abolished cold-induced BAT oxidative activity and impaired FA metabolism in BAHKO mice. Our findings show that brown adipocyte intracellular lipolysis is required for BAT thermogenesis.
Collapse
Affiliation(s)
- Etienne Mouisel
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France.
| | - Anaïs Bodon
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Christophe Noll
- Centre de recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada; Department of Medicine, Division of Endocrinology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Stéphanie Cassant-Sourdy
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Marie-Adeline Marques
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Remy Flores-Flores
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Elodie Riant
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Camille Bergoglio
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Pierre Vezin
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Sylvie Caspar-Bauguil
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France; Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Camille Fournes-Fraresso
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Geneviève Tavernier
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Khalil Acheikh Ibn Oumar
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Pierre Gourdy
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France; Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Denis P Blondin
- Centre de recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada; Department of Medicine, Division of Neurology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Pierre-Damien Denechaud
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - André C Carpentier
- Centre de recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada; Department of Medicine, Division of Endocrinology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Dominique Langin
- Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, Université de Toulouse, Inserm, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France; Centre Hospitalier Universitaire de Toulouse, Toulouse, France; Institut Universitaire de France (IUF), Paris, France.
| |
Collapse
|
3
|
Katayama M, Nomura K, Mudry JM, Chibalin AV, Krook A, Zierath JR. Exercise-induced methylation of the Serhl2 promoter and implication for lipid metabolism in rat skeletal muscle. Mol Metab 2025; 92:102081. [PMID: 39657853 PMCID: PMC11732562 DOI: 10.1016/j.molmet.2024.102081] [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: 10/01/2024] [Revised: 11/22/2024] [Accepted: 12/03/2024] [Indexed: 12/12/2024] Open
Abstract
OBJECTIVES Environmental factors such as physical activity induce epigenetic modifications, with exercise-responsive DNA methylation changes occurring in skeletal muscle. To determine the skeletal muscle DNA methylation signature of endurance swim training, we used whole-genome methylated DNA immunoprecipitation (MeDIP) sequencing. METHODS We utilized endurance-trained rats, cultured L6 myotubes, and human skeletal muscle cells, employing MeDIP sequencing, gene silencing, and palmitate oxidation assays. Additional methods included promoter luciferase assays, fluorescence microscopy, and RNA/DNA analysis to investigate exercise-induced molecular changes. RESULTS Gene set enrichment analysis (GSEA) of differentially methylated promoter regions identified an enrichment of four gene sets, including those linked to lipid metabolic processes, with hypermethylated or hypomethylated promoter regions in skeletal muscle of exercise-trained rats. Bisulfite sequencing confirmed hypomethylation of CpGs in the Serhl2 (Serine Hydrolase Like 2) transcription start site in exercise-trained rats. Serhl2 gene expression was upregulated in both exercise-trained rats and an "exercise-in-a-dish" model of L6 myotubes subjected to electrical pulse stimulation (EPS). Serhl2 promoter activity was regulated by methylation and EPS. A Nr4a binding motif in the Serhl2 promoter, when deleted, reduced promoter activity and sensitivity to methylation in L6 myotubes. Silencing Serhl2 in L6 myotubes reduced intracellular lipid oxidation and triacylglycerol synthesis in response to EPS. CONCLUSIONS Exercise-training enhances intracellular lipid metabolism and phenotypic changes in skeletal muscle through epigenomic modifications on Serhl2. Hypomethylation of the Serhl2 promoter influences Nr4a transcription factor binding, promoter activity, and gene expression, linking exercise-induced epigenomic regulation of Serhl2 to lipid oxidation and triacylglycerol synthesis.
Collapse
Affiliation(s)
- Mutsumi Katayama
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Kazuhiro Nomura
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Jonathan M Mudry
- Section of Integrative Physiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Alexander V Chibalin
- Section of Integrative Physiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Anna Krook
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Juleen R Zierath
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Section of Integrative Physiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
4
|
Sakamoto K, Butera MA, Zhou C, Maurizi G, Chen B, Ling L, Shawkat A, Patlolla L, Thakker K, Calle V, Morgan DA, Rahmouni K, Schwartz GJ, Tahiri A, Buettner C. Overnutrition causes insulin resistance and metabolic disorder through increased sympathetic nervous system activity. Cell Metab 2025; 37:121-137.e6. [PMID: 39437790 PMCID: PMC11711004 DOI: 10.1016/j.cmet.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/19/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
The mechanisms underlying obesity-induced insulin resistance remain incompletely understood, as impaired cellular insulin signaling, traditionally considered the primary driver of insulin resistance, does not always accompany impaired insulin action. Overnutrition rapidly increases plasma norepinephrine (NE), suggesting overactivation of the sympathetic nervous system (SNS). However, the role of the SNS in obesity is controversial, as both increased and decreased SNS activity (SNA) have been reported. Here, we show that reducing catecholamine (CA) release from the SNS protects against overnutrition-induced insulin resistance as well as hyperglucagonemia, adipose tissue dysfunction, and fatty liver disease, as we demonstrate utilizing a mouse model of inducible and peripherally restricted deletion of tyrosine hydroxylase (th; THΔper). A key mechanism through which heightened SNA induces insulin resistance is by triggering adipose tissue lipolysis. Increased SNA emerges as a critical driver in the pathogenesis of overnutrition-induced insulin resistance and metabolic disease independent of cellular insulin signaling.
Collapse
Affiliation(s)
- Kenichi Sakamoto
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary A Butera
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chunxue Zhou
- Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giulia Maurizi
- Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bandy Chen
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Ling
- Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adham Shawkat
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Likhitha Patlolla
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Kavira Thakker
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Victor Calle
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Donald A Morgan
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Gary J Schwartz
- Department of Medicine & Neuroscience, Albert Einstein College of Medicine, New York, NY, USA
| | - Azeddine Tahiri
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Christoph Buettner
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
5
|
Ding L, Huwyler F, Long F, Yang W, Binz J, Wernlé K, Pfister M, Klug M, Balaz M, Ukropcova B, Ukropec J, Wu C, Wang T, Gao M, Clavien PA, Dutkowski P, Tibbitt MW, Wolfrum C. Glucose controls lipolysis through Golgi PtdIns4P-mediated regulation of ATGL. Nat Cell Biol 2024; 26:552-566. [PMID: 38561547 PMCID: PMC11021197 DOI: 10.1038/s41556-024-01386-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 02/23/2024] [Indexed: 04/04/2024]
Abstract
Metabolic crosstalk of the major nutrients glucose, amino acids and fatty acids (FAs) ensures systemic metabolic homeostasis. The coordination between the supply of glucose and FAs to meet various physiological demands is especially important as improper nutrient levels lead to metabolic disorders, such as diabetes and metabolic dysfunction-associated steatohepatitis (MASH). In response to the oscillations in blood glucose levels, lipolysis is thought to be mainly regulated hormonally to control FA liberation from lipid droplets by insulin, catecholamine and glucagon. However, whether general cell-intrinsic mechanisms exist to directly modulate lipolysis via glucose sensing remains largely unknown. Here we report the identification of such an intrinsic mechanism, which involves Golgi PtdIns4P-mediated regulation of adipose triglyceride lipase (ATGL)-driven lipolysis via intracellular glucose sensing. Mechanistically, depletion of intracellular glucose results in lower Golgi PtdIns4P levels, and thus reduced assembly of the E3 ligase complex CUL7FBXW8 in the Golgi apparatus. Decreased levels of the E3 ligase complex lead to reduced polyubiquitylation of ATGL in the Golgi and enhancement of ATGL-driven lipolysis. This cell-intrinsic mechanism regulates both the pool of intracellular FAs and their extracellular release to meet physiological demands during fasting and glucose deprivation. Moreover, genetic and pharmacological manipulation of the Golgi PtdIns4P-CUL7FBXW8-ATGL axis in mouse models of simple hepatic steatosis and MASH, as well as during ex vivo perfusion of a human steatotic liver graft leads to the amelioration of steatosis, suggesting that this pathway might be a promising target for metabolic dysfunction-associated steatotic liver disease and possibly MASH.
Collapse
Affiliation(s)
- Lianggong Ding
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
| | - Florian Huwyler
- Macromolecular Engineering Laboratory, Institute of Energy and Process Engineering, ETH Zürich, Zurich, Switzerland
| | - Fen Long
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
| | - Wu Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Jonas Binz
- Macromolecular Engineering Laboratory, Institute of Energy and Process Engineering, ETH Zürich, Zurich, Switzerland
| | - Kendra Wernlé
- Department of Surgery and Transplantation, University of Zurich, Zurich, Switzerland
- Wyss Zurich Translational Center, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Matthias Pfister
- Department of Surgery and Transplantation, University of Zurich, Zurich, Switzerland
- Wyss Zurich Translational Center, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Manuel Klug
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
| | - Miroslav Balaz
- Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Barbara Ukropcova
- Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Jozef Ukropec
- Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Chunyan Wu
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
| | - Tongtong Wang
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
| | - Min Gao
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
- Department of Pharmacy, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Pierre-Alain Clavien
- Department of Surgery and Transplantation, University of Zurich, Zurich, Switzerland
- Wyss Zurich Translational Center, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Philipp Dutkowski
- Department of Surgery and Transplantation, University of Zurich, Zurich, Switzerland
| | - Mark W Tibbitt
- Macromolecular Engineering Laboratory, Institute of Energy and Process Engineering, ETH Zürich, Zurich, Switzerland
- Wyss Zurich Translational Center, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland.
| |
Collapse
|
6
|
Fernandes MF, Aristizabal-Henao JJ, Marvyn PM, M'Hiri I, Wiens MA, Hoang M, Sebastian M, Nachbar R, St-Pierre P, Diaguarachchige De Silva K, Wood GA, Joseph JW, Doucette CA, Marette A, Stark KD, Duncan RE. Renal tubule-specific Atgl deletion links kidney lipid metabolism to glucagon-like peptide 1 and insulin secretion independent of renal inflammation or lipotoxicity. Mol Metab 2024; 81:101887. [PMID: 38280449 PMCID: PMC10850971 DOI: 10.1016/j.molmet.2024.101887] [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: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 01/29/2024] Open
Abstract
OBJECTIVE Lipotoxic injury from renal lipid accumulation in obesity and type 2 diabetes (T2D) is implicated in associated kidney damage. However, models examining effects of renal ectopic lipid accumulation independent of obesity or T2D are lacking. We generated renal tubule-specific adipose triglyceride lipase knockout (RT-SAKO) mice to determine if this targeted triacylglycerol (TAG) over-storage affects glycemic control and kidney health. METHODS Male and female RT-SAKO mice and their control littermates were tested for changes in glycemic control at 10-12 and 16-18 weeks of age. Markers of kidney health and blood lipid and hormone concentrations were analyzed. Kidney and blood lysophosphatidic acid (LPA) levels were measured, and a role for LPA in mediating impaired glycemic control was evaluated using the LPA receptor 1/3 inhibitor Ki-16425. RESULTS All groups remained insulin sensitive, but 16- to 18-week-old male RT-SAKO mice became glucose intolerant, without developing kidney inflammation or fibrosis. Rather, these mice displayed lower circulating insulin and glucagon-like peptide 1 (GLP-1) levels. Impaired first-phase glucose-stimulated insulin secretion was detected and restored by Exendin-4. Kidney and blood LPA levels were elevated in older male but not female RT-SAKO mice, associated with increased kidney diacylglycerol kinase epsilon. Inhibition of LPA-mediated signaling restored serum GLP-1 levels, first-phase insulin secretion, and glucose tolerance. CONCLUSIONS TAG over-storage alone is insufficient to cause renal tubule lipotoxicity. This work is the first to show that endogenously derived LPA modulates GLP-1 levels in vivo, demonstrating a new mechanism of kidney-gut-pancreas crosstalk to regulate insulin secretion and glucose homeostasis.
Collapse
Affiliation(s)
- Maria F Fernandes
- Department of Kinesiology and Health Sciences, University of Waterloo, Ontario, Canada
| | | | - Phillip M Marvyn
- Department of Kinesiology and Health Sciences, University of Waterloo, Ontario, Canada
| | - Iman M'Hiri
- Department of Kinesiology and Health Sciences, University of Waterloo, Ontario, Canada
| | - Meghan A Wiens
- Department of Kinesiology and Health Sciences, University of Waterloo, Ontario, Canada
| | - Monica Hoang
- School of Pharmacy, University of Waterloo, Ontario, Canada
| | - Manuel Sebastian
- Max Rady College of Medicine, University of Manitoba, Manitoba, Canada
| | - Renato Nachbar
- Québec Heart and Lung Institute, Department of Medicine, Laval University, Québec, Canada
| | - Philippe St-Pierre
- Québec Heart and Lung Institute, Department of Medicine, Laval University, Québec, Canada
| | | | - Geoffrey A Wood
- Ontario Veterinary College, University of Guelph, Ontario, Canada
| | - Jamie W Joseph
- School of Pharmacy, University of Waterloo, Ontario, Canada
| | | | - André Marette
- Québec Heart and Lung Institute, Department of Medicine, Laval University, Québec, Canada
| | - Ken D Stark
- Department of Kinesiology and Health Sciences, University of Waterloo, Ontario, Canada
| | - Robin E Duncan
- Department of Kinesiology and Health Sciences, University of Waterloo, Ontario, Canada.
| |
Collapse
|
7
|
Gehle PF, Satzinger S, Willenborg S, Eming SA. Isolation of dermal adipocytes from mouse skin for biochemical analysis. STAR Protoc 2023; 4:102765. [PMID: 38060383 PMCID: PMC10751568 DOI: 10.1016/j.xpro.2023.102765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 10/04/2023] [Accepted: 11/21/2023] [Indexed: 12/30/2023] Open
Abstract
The role of dermal white adipose tissue in regulating skin homeostasis and self-renewal processes has recently attracted interest. However, the isolation of proteins from dermal adipocytes for biochemical analysis is challenging. Here, we provide a protocol for the isolation of murine dermal adipocytes. We describe steps for inducing adipocyte-specific gene deletion, adipocyte isolation, protein purification, and western blot analysis. The reliability of the protocol is demonstrated by verifying efficient adipocyte-specific Atgl gene deletion in a tamoxifen-inducible Cre/loxP-based mouse model. For complete details on the use and execution of this protocol, please refer to Zhang et al. (2019).1.
Collapse
Affiliation(s)
- Paul F Gehle
- Department of Dermatology, University of Cologne, 50937 Cologne, Germany.
| | - Sabrina Satzinger
- Department of Dermatology, University of Cologne, 50937 Cologne, Germany
| | | | - Sabine A Eming
- Department of Dermatology, University of Cologne, 50937 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Institute of Zoology, Developmental Biology Unit, University of Cologne, 50674 Cologne, Germany.
| |
Collapse
|
8
|
Nandy A, Helderman RCM, Thapa S, Jayapalan S, Richards A, Narayani N, Czech MP, Rosen CJ, Rendina-Ruedy E. Lipolysis supports bone formation by providing osteoblasts with endogenous fatty acid substrates to maintain bioenergetic status. Bone Res 2023; 11:62. [PMID: 38001111 PMCID: PMC10673934 DOI: 10.1038/s41413-023-00297-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/18/2023] [Accepted: 09/22/2023] [Indexed: 11/26/2023] Open
Abstract
Bone formation is a highly energy-demanding process that can be impacted by metabolic disorders. Glucose has been considered the principal substrate for osteoblasts, although fatty acids are also important for osteoblast function. Here, we report that osteoblasts can derive energy from endogenous fatty acids stored in lipid droplets via lipolysis and that this process is critical for bone formation. As such, we demonstrate that osteoblasts accumulate lipid droplets that are highly dynamic and provide the molecular mechanism by which they serve as a fuel source for energy generation during osteoblast maturation. Inhibiting cytoplasmic lipolysis leads to both an increase in lipid droplet size in osteoblasts and an impairment in osteoblast function. The fatty acids released by lipolysis from these lipid droplets become critical for cellular energy production as cellular energetics shifts towards oxidative phosphorylation during nutrient-depleted conditions. In vivo, conditional deletion of the ATGL-encoding gene Pnpla2 in osteoblast progenitor cells reduces cortical and trabecular bone parameters and alters skeletal lipid metabolism. Collectively, our data demonstrate that osteoblasts store fatty acids in the form of lipid droplets, which are released via lipolysis to support cellular bioenergetic status when nutrients are limited. Perturbations in this process result in impairment of bone formation, specifically reducing ATP production and overall osteoblast function.
Collapse
Affiliation(s)
- Ananya Nandy
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Ron C M Helderman
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Santosh Thapa
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Shobana Jayapalan
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Alison Richards
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Nikita Narayani
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | | | - Elizabeth Rendina-Ruedy
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
9
|
Machado H, Hofer P, Zechner R, Smith TK, Figueiredo LM. Adipocyte lipolysis protects mice against Trypanosoma brucei infection. Nat Microbiol 2023; 8:2020-2032. [PMID: 37828246 PMCID: PMC10627827 DOI: 10.1038/s41564-023-01496-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 09/11/2023] [Indexed: 10/14/2023]
Abstract
Trypanosoma brucei causes African trypanosomiasis, colonizing adipose tissue and inducing weight loss. Here we investigated the molecular mechanisms responsible for adipose mass loss and its impact on disease pathology. We found that lipolysis is activated early in infection. Mice lacking B and T lymphocytes fail to upregulate adipocyte lipolysis, resulting in higher fat mass retention. Genetic ablation of the rate-limiting adipose triglyceride lipase specifically from adipocytes (AdipoqCre/+-Atglfl/fl) prevented the stimulation of adipocyte lipolysis during infection, reducing fat mass loss. Surprisingly, these mice succumbed earlier and presented a higher parasite burden in the gonadal adipose tissue, indicating that host lipolysis limits parasite growth. Consistently, free fatty acids comparable with those of adipose interstitial fluid induced loss of parasite viability. Adipocyte lipolysis emerges as a mechanism controlling local parasite burden and affecting the loss of fat mass in African trypanosomiasis.
Collapse
Affiliation(s)
- Henrique Machado
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Peter Hofer
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Terry K Smith
- School of Biology, Biomedical Sciences Research Complex, University of St Andrews, St Andrews, UK
| | - Luísa M Figueiredo
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
10
|
Kim KH, Jia Z, Snyder M, Chen J, Qiu J, Oprescu SN, Chen X, Syed SA, Yue F, Roseguini BT, Imbalzano AN, Hu C, Kuang S. PRMT5 links lipid metabolism to contractile function of skeletal muscles. EMBO Rep 2023; 24:e57306. [PMID: 37334900 PMCID: PMC10398672 DOI: 10.15252/embr.202357306] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/30/2023] [Accepted: 06/05/2023] [Indexed: 06/21/2023] Open
Abstract
Skeletal muscle plays a key role in systemic energy homeostasis besides its contractile function, but what links these functions is poorly defined. Protein Arginine Methyl Transferase 5 (PRMT5) is a well-known oncoprotein but also expressed in healthy tissues with unclear physiological functions. As adult muscles express high levels of Prmt5, we generated skeletal muscle-specific Prmt5 knockout (Prmt5MKO ) mice. We observe reduced muscle mass, oxidative capacity, force production, and exercise performance in Prmt5MKO mice. The motor deficiency is associated with scarce lipid droplets in myofibers due to defects in lipid biosynthesis and accelerated degradation. Specifically, PRMT5 deletion reduces dimethylation and stability of Sterol Regulatory Element-Binding Transcription Factor 1a (SREBP1a), a master regulator of de novo lipogenesis. Moreover, Prmt5MKO impairs the repressive H4R3 symmetric dimethylation at the Pnpla2 promoter, elevating the level of its encoded protein ATGL, the rate-limiting enzyme catalyzing lipolysis. Accordingly, skeletal muscle-specific double knockout of Pnpla2 and Prmt5 normalizes muscle mass and function. Together, our findings delineate a physiological function of PRMT5 in linking lipid metabolism to contractile function of myofibers.
Collapse
Affiliation(s)
- Kun Ho Kim
- Department of Animal SciencesPurdue UniversityWest LafayetteINUSA
| | - Zhihao Jia
- Department of Animal SciencesPurdue UniversityWest LafayetteINUSA
| | - Madigan Snyder
- Department of Animal SciencesPurdue UniversityWest LafayetteINUSA
- Department of Biological SciencesPurdue UniversityWest LafayetteINUSA
| | - Jingjuan Chen
- Department of Animal SciencesPurdue UniversityWest LafayetteINUSA
| | - Jiamin Qiu
- Department of Animal SciencesPurdue UniversityWest LafayetteINUSA
| | - Stephanie N Oprescu
- Department of Animal SciencesPurdue UniversityWest LafayetteINUSA
- Department of Biological SciencesPurdue UniversityWest LafayetteINUSA
| | - Xiyue Chen
- Department of Animal SciencesPurdue UniversityWest LafayetteINUSA
| | - Sabriya A Syed
- Department of Biochemistry and Molecular PharmacologyUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Feng Yue
- Department of Animal SciencesPurdue UniversityWest LafayetteINUSA
| | - Bruno T Roseguini
- Department of Health and KinesiologyPurdue UniversityWest LafayetteINUSA
| | - Anthony N Imbalzano
- Department of Biochemistry and Molecular PharmacologyUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Changdeng Hu
- Department of Medicinal Chemistry and Molecular PharmacologyPurdue UniversityWest LafayetteINUSA
- Center for Cancer ResearchPurdue UniversityWest LafayetteINUSA
| | - Shihuan Kuang
- Department of Animal SciencesPurdue UniversityWest LafayetteINUSA
- Center for Cancer ResearchPurdue UniversityWest LafayetteINUSA
| |
Collapse
|
11
|
Alekos NS, Kushwaha P, Kim SP, Li Z, Abood A, Dirckx N, Aja S, Kodama J, Garcia-Diaz JG, Otsuru S, Rendina-Ruedy E, Wolfgang MJ, Riddle RC. Mitochondrial β-oxidation of adipose-derived fatty acids by osteoblasts fuels parathyroid hormone-induced bone formation. JCI Insight 2023; 8:e165604. [PMID: 36729662 PMCID: PMC10070112 DOI: 10.1172/jci.insight.165604] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 02/01/2023] [Indexed: 02/03/2023] Open
Abstract
The energetic costs of bone formation require osteoblasts to coordinate their activities with tissues, like adipose, that can supply energy-dense macronutrients. In the case of intermittent parathyroid hormone (PTH) treatment, a strategy used to reduce fracture risk, bone formation is preceded by a change in systemic lipid homeostasis. To investigate the requirement for fatty acid oxidation by osteoblasts during PTH-induced bone formation, we subjected mice with osteoblast-specific deficiency of mitochondrial long-chain β-oxidation as well as mice with adipocyte-specific deficiency for the PTH receptor or adipose triglyceride lipase to an anabolic treatment regimen. PTH increased the release of fatty acids from adipocytes and β-oxidation by osteoblasts, while the genetic mouse models were resistant to the hormone's anabolic effect. Collectively, these data suggest that PTH's anabolic actions require coordinated signaling between bone and adipose, wherein a lipolytic response liberates fatty acids that are oxidized by osteoblasts to fuel bone formation.
Collapse
Affiliation(s)
- Nathalie S. Alekos
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Priyanka Kushwaha
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Soohyun P. Kim
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zhu Li
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Abdullah Abood
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Naomi Dirckx
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Susan Aja
- Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Joe Kodama
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jean G. Garcia-Diaz
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Satoru Otsuru
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Elizabeth Rendina-Ruedy
- Department of Medicine and Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michael J. Wolfgang
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ryan C. Riddle
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Research & Development Service, Baltimore Veterans Administration Medical Center, Baltimore, Maryland, USA
| |
Collapse
|
12
|
Richter FC, Friedrich M, Kampschulte N, Piletic K, Alsaleh G, Zummach R, Hecker J, Pohin M, Ilott N, Guschina I, Wideman SK, Johnson E, Borsa M, Hahn P, Morriseau C, Hammock BD, Schipper HS, Edwards CM, Zechner R, Siegmund B, Weidinger C, Schebb NH, Powrie F, Simon AK. Adipocyte autophagy limits gut inflammation by controlling oxylipin and IL-10. EMBO J 2023; 42:e112202. [PMID: 36795015 PMCID: PMC10015370 DOI: 10.15252/embj.2022112202] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 02/17/2023] Open
Abstract
Lipids play a major role in inflammatory diseases by altering inflammatory cell functions, either through their function as energy substrates or as lipid mediators such as oxylipins. Autophagy, a lysosomal degradation pathway that limits inflammation, is known to impact on lipid availability, however, whether this controls inflammation remains unexplored. We found that upon intestinal inflammation visceral adipocytes upregulate autophagy and that adipocyte-specific loss of the autophagy gene Atg7 exacerbates inflammation. While autophagy decreased lipolytic release of free fatty acids, loss of the major lipolytic enzyme Pnpla2/Atgl in adipocytes did not alter intestinal inflammation, ruling out free fatty acids as anti-inflammatory energy substrates. Instead, Atg7-deficient adipose tissues exhibited an oxylipin imbalance, driven through an NRF2-mediated upregulation of Ephx1. This shift reduced secretion of IL-10 from adipose tissues, which was dependent on the cytochrome P450-EPHX pathway, and lowered circulating levels of IL-10 to exacerbate intestinal inflammation. These results suggest an underappreciated fat-gut crosstalk through an autophagy-dependent regulation of anti-inflammatory oxylipins via the cytochrome P450-EPHX pathway, indicating a protective effect of adipose tissues for distant inflammation.
Collapse
Affiliation(s)
| | - Matthias Friedrich
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
- Translational Gastroenterology Unit, Nuffield Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Nadja Kampschulte
- Faculty of Mathematics and Natural SciencesUniversity of WuppertalWuppertalGermany
| | - Klara Piletic
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Ghada Alsaleh
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | | | - Julia Hecker
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin and Berlin Institute of HealthBerlinGermany
- Department of Gastroenterology, Infectious Diseases and RheumatologyCampus Benjamin FranklinBerlinGermany
| | - Mathilde Pohin
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Nicholas Ilott
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | | | - Sarah Karin Wideman
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Errin Johnson
- The Dunn School of PathologyUniversity of OxfordOxfordUK
| | - Mariana Borsa
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Paula Hahn
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Christophe Morriseau
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer CenterUniversity of CaliforniaDavisCAUSA
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer CenterUniversity of CaliforniaDavisCAUSA
| | - Henk Simon Schipper
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
- Center for Translational ImmunologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Claire M Edwards
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research CentreUniversity of OxfordOxfordUK
- Nuffield Department of Surgical Sciences, Botnar Research CentreUniversity of OxfordOxfordUK
| | - Rudolf Zechner
- Institute of Molecular BiosciencesUniversity of GrazGrazAustria
| | - Britta Siegmund
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin and Berlin Institute of HealthBerlinGermany
- Department of Gastroenterology, Infectious Diseases and RheumatologyCampus Benjamin FranklinBerlinGermany
| | - Carl Weidinger
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin and Berlin Institute of HealthBerlinGermany
- Department of Gastroenterology, Infectious Diseases and RheumatologyCampus Benjamin FranklinBerlinGermany
| | - Nils Helge Schebb
- Faculty of Mathematics and Natural SciencesUniversity of WuppertalWuppertalGermany
| | - Fiona Powrie
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Anna Katharina Simon
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
- Max Delbrück CenterBerlinGermany
| |
Collapse
|
13
|
Bielawiec P, Dziemitko S, Konstantynowicz-Nowicka K, Chabowski A, Dzięcioł J, Harasim-Symbor E. Cannabidiol improves muscular lipid profile by affecting the expression of fatty acid transporters and inhibiting de novo lipogenesis. Sci Rep 2023; 13:3694. [PMID: 36879113 PMCID: PMC9988888 DOI: 10.1038/s41598-023-30872-w] [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: 12/09/2022] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Obesity is one of the principal public health concerns leading to disturbances in glucose and lipid metabolism, which is a risk factor for several chronic diseases, including insulin resistance, type 2 diabetes mellitus, and cardiovascular diseases. In recent years, it turned out that cannabidiol (CBD) is a potential therapeutic agent in the treatment of obesity and its complications. Therefore, in the present study, we used CBD therapy (intraperitoneal injections in a dose of 10 mg/kg of body mass for 14 days) in a rat model of obesity induced by a high-fat diet (HFD). Gas-liquid chromatography and Western blotting were applied in order to determine the intramuscular lipid content and total expression of selected proteins in the white and red gastrocnemius muscle, respectively. Based on fatty acid composition, we calculated de novo lipogenesis ratio (16:0/18:2n-6), desaturation ratio (18:1n-9/18:0), and elongation ratios (18:0/16:0, 20:0/18:0, 22:0/20:0 and 24:0/22:0), in the selected lipid fractions. Two-week CBD administration significantly reduced the intramuscular fatty acids (FAs) accumulation and inhibited de novo lipogenesis in different lipid pools (in the free fatty acid, diacylglycerol, and triacylglycerol fractions) in both muscle types, which coincided with a decrease in the expression of membrane fatty acid transporters (fatty acid translocase, membrane-associated fatty acid binding protein, and fatty acid transport proteins 1 and 4). Moreover, CBD application profoundly improved the elongation and desaturation ratios, which was in line with downregulated expression of enzymes from the family of elongases and desaturases regardless of the metabolism presented by the muscle type. To our knowledge, this study is the first that outlines the novel effects of CBD action on skeletal muscle with different types of metabolism (oxidative vs. glycolytic).
Collapse
Affiliation(s)
- Patrycja Bielawiec
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland.
| | - Sylwia Dziemitko
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | | | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Janusz Dzięcioł
- Department of Human Anatomy, Medical University of Bialystok, Bialystok, Poland
| | - Ewa Harasim-Symbor
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
14
|
Chronic intake of high dietary sucrose induces sexually dimorphic metabolic adaptations in mouse liver and adipose tissue. Nat Commun 2022; 13:6062. [PMID: 36229459 PMCID: PMC9561177 DOI: 10.1038/s41467-022-33840-6] [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] [Received: 06/29/2020] [Accepted: 10/05/2022] [Indexed: 01/05/2023] Open
Abstract
Almost all effective treatments for non-alcoholic fatty liver disease (NAFLD) involve reduction of adiposity, which suggests the metabolic axis between liver and adipose tissue is essential to NAFLD development. Since excessive dietary sugar intake may be an initiating factor for NAFLD, we have characterized the metabolic effects of liquid sucrose intake at concentrations relevant to typical human consumption in mice. We report that sucrose intake induces sexually dimorphic effects in liver, adipose tissue, and the microbiome; differences concordant with steatosis severity. We show that when steatosis is decoupled from impairments in insulin responsiveness, sex is a moderating factor that influences sucrose-driven lipid storage and the contribution of de novo fatty acid synthesis to the overall hepatic triglyceride pool. Our findings provide physiologic insight into how sex influences the regulation of adipose-liver crosstalk and highlight the importance of extrahepatic metabolism in the pathogenesis of diet-induced steatosis and NAFLD.
Collapse
|
15
|
Fuchs CD, Radun R, Dixon ED, Mlitz V, Timelthaler G, Halilbasic E, Herac M, Jonker JW, Ronda OAHO, Tardelli M, Haemmerle G, Zimmermann R, Scharnagl H, Stojakovic T, Verkade HJ, Trauner M. Hepatocyte-specific deletion of adipose triglyceride lipase (adipose triglyceride lipase/patatin-like phospholipase domain containing 2) ameliorates dietary induced steatohepatitis in mice. Hepatology 2022; 75:125-139. [PMID: 34387896 DOI: 10.1002/hep.32112] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/29/2021] [Accepted: 08/11/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIMS Increased fatty acid (FA) flux from adipose tissue to the liver contributes to the development of NAFLD. Because free FAs are key lipotoxic triggers accelerating disease progression, inhibiting adipose triglyceride lipase (ATGL)/patatin-like phospholipase domain containing 2 (PNPLA2), the main enzyme driving lipolysis, may attenuate steatohepatitis. APPROACH AND RESULTS Hepatocyte-specific ATGL knockout (ATGL LKO) mice were challenged with methionine-choline-deficient (MCD) or high-fat high-carbohydrate (HFHC) diet. Serum biochemistry, hepatic lipid content and liver histology were assessed. Mechanistically, hepatic gene and protein expression of lipid metabolism, inflammation, fibrosis, apoptosis, and endoplasmic reticulum (ER) stress markers were investigated. DNA binding activity for peroxisome proliferator-activated receptor (PPAR) α and PPARδ was measured. After short hairpin RNA-mediated ATGL knockdown, HepG2 cells were treated with lipopolysaccharide (LPS) or oleic acid:palmitic acid 2:1 (OP21) to explore the direct role of ATGL in inflammation in vitro. On MCD and HFHC challenge, ATGL LKO mice showed reduced PPARα and increased PPARδ DNA binding activity when compared with challenged wild-type (WT) mice. Despite histologically and biochemically pronounced hepatic steatosis, dietary-challenged ATGL LKO mice showed lower hepatic inflammation, reflected by the reduced number of Galectin3/MAC-2 and myeloperoxidase-positive cells and low mRNA expression levels of inflammatory markers (such as IL-1β and F4/80) when compared with WT mice. In line with this, protein levels of the ER stress markers protein kinase R-like endoplasmic reticulum kinase and inositol-requiring enzyme 1α were reduced in ATGL LKO mice fed with MCD diet. Accordingly, pretreatment of LPS-treated HepG2 cells with the PPARδ agonist GW0742 suppressed mRNA expression of inflammatory markers. Additionally, ATGL knockdown in HepG2 cells attenuated LPS/OP21-induced expression of proinflammatory cytokines and chemokines such as chemokine (C-X-C motif) ligand 5, chemokine (C-C motif) ligand (Ccl) 2, and Ccl5. CONCLUSIONS Low hepatic lipolysis and increased PPARδ activity in ATGL/PNPLA2 deficiency may counteract hepatic inflammation and ER stress despite increased steatosis. Therefore, lowering hepatocyte lipolysis through ATGL inhibition represents a promising therapeutic strategy for the treatment of steatohepatitis.
Collapse
Affiliation(s)
- Claudia D Fuchs
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Richard Radun
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Emmanuel D Dixon
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Veronika Mlitz
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Gerald Timelthaler
- Institute for Cancer Research, Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Emina Halilbasic
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Merima Herac
- Clinical Institute of Pathology, Medical University Vienna, Vienna, Austria
| | - Johan W Jonker
- Department of Pediatrics, Section of Molecular Metabolism and Nutrition, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Onne A H O Ronda
- Pediatric Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Matteo Tardelli
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria.,Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Guenter Haemmerle
- BioTechMed-Graz, Graz, Austria.,Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Robert Zimmermann
- BioTechMed-Graz, Graz, Austria.,Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Tatjana Stojakovic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, University Hospital Graz, Graz, Austria
| | - Henkjan J Verkade
- Pediatric Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
16
|
Ding L, Sun W, Balaz M, He A, Klug M, Wieland S, Caiazzo R, Raverdy V, Pattou F, Lefebvre P, Lodhi IJ, Staels B, Heim M, Wolfrum C. Peroxisomal β-oxidation acts as a sensor for intracellular fatty acids and regulates lipolysis. Nat Metab 2021; 3:1648-1661. [PMID: 34903883 PMCID: PMC8688145 DOI: 10.1038/s42255-021-00489-2] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 10/06/2021] [Indexed: 01/07/2023]
Abstract
To liberate fatty acids (FAs) from intracellular stores, lipolysis is regulated by the activity of the lipases adipose triglyceride lipase (ATGL), hormone-sensitive lipase and monoacylglycerol lipase. Excessive FA release as a result of uncontrolled lipolysis results in lipotoxicity, which can in turn promote the progression of metabolic disorders. However, whether cells can directly sense FAs to maintain cellular lipid homeostasis is unknown. Here we report a sensing mechanism for cellular FAs based on peroxisomal degradation of FAs and coupled with reactive oxygen species (ROS) production, which in turn regulates FA release by modulating lipolysis. Changes in ROS levels are sensed by PEX2, which modulates ATGL levels through post-translational ubiquitination. We demonstrate the importance of this pathway for non-alcoholic fatty liver disease progression using genetic and pharmacological approaches to alter ROS levels in vivo, which can be utilized to increase hepatic ATGL levels and ameliorate hepatic steatosis. The discovery of this peroxisomal β-oxidation-mediated feedback mechanism, which is conserved in multiple organs, couples the functions of peroxisomes and lipid droplets and might serve as a new way to manipulate lipolysis to treat metabolic disorders.
Collapse
Affiliation(s)
- Lianggong Ding
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
| | - Wenfei Sun
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
| | - Miroslav Balaz
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
- Institute of Experimental Endocrinology, Biomedical Research Center at the Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia
| | - Anyuan He
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- School of Life Sciences, Anhui Medical University, Hefei, China
| | - Manuel Klug
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland
| | - Stefan Wieland
- Hepatology, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Robert Caiazzo
- University Lille, CHU Lille, Institut Pasteur Lille, Inserm, UMR1190 Translational Research in Diabetes, Lille, France
| | - Violeta Raverdy
- University Lille, CHU Lille, Institut Pasteur Lille, Inserm, UMR1190 Translational Research in Diabetes, Lille, France
| | - Francois Pattou
- University Lille, CHU Lille, Institut Pasteur Lille, Inserm, UMR1190 Translational Research in Diabetes, Lille, France
| | - Philippe Lefebvre
- University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Bart Staels
- University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Markus Heim
- Hepatology, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
- Division of Gastroenterology and Hepatology, Clarunis, University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach, Switzerland.
| |
Collapse
|
17
|
Kakehi S, Tamura Y, Ikeda SI, Kaga N, Taka H, Ueno N, Shiuchi T, Kubota A, Sakuraba K, Kawamori R, Watada H. Short-term physical inactivity induces diacylglycerol accumulation and insulin resistance in muscle via lipin1 activation. Am J Physiol Endocrinol Metab 2021; 321:E766-E781. [PMID: 34719943 DOI: 10.1152/ajpendo.00254.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Physical inactivity impairs muscle insulin sensitivity. However, its mechanism is unclear. To model physical inactivity, we applied 24-h hind-limb cast immobilization (HCI) to mice with normal or high-fat diet (HFD) and evaluated intramyocellular lipids and the insulin signaling pathway in the soleus muscle. Although 2-wk HFD alone did not alter intramyocellular diacylglycerol (IMDG) accumulation, HCI alone increased it by 1.9-fold and HCI after HFD further increased it by 3.3-fold. Parallel to this, we found increased protein kinase C ε (PKCε) activity, reduced insulin-induced 2-deoxyglucose (2-DOG) uptake, and reduced phosphorylation of insulin receptor β (IRβ) and Akt, key molecules for insulin signaling pathway. Lipin1, which converts phosphatidic acid to diacylglycerol, showed increase of its activity by HCI, and dominant-negative lipin1 expression in muscle prevented HCI-induced IMDG accumulation and impaired insulin-induced 2-DOG uptake. Furthermore, 24-h leg cast immobilization in human increased lipin1 expression. Thus, even short-term immobilization increases IMDG and impairs insulin sensitivity in muscle via enhanced lipin1 activity.NEW & NOTEWORTHY Physical inactivity impairs muscle insulin sensitivity. However, its mechanism is unclear. To model physical inactivity, we applied 24-h hind-limb cast immobilization to mice with normal or high-fat diet and evaluated intramyocellular lipids and the insulin signaling pathway in the soleus muscle. We found that even short-term immobilization increases intramyocellular diacylglycerol and impairs insulin sensitivity in muscle via enhanced lipin1 activity.
Collapse
Affiliation(s)
- Saori Kakehi
- Department of Metabolism and Endocrinology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Sportology Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yoshifumi Tamura
- Department of Metabolism and Endocrinology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Sportology Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Shin-Ichi Ikeda
- Department of Metabolism and Endocrinology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Sportology Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Naoko Kaga
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Hikari Taka
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Noriko Ueno
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Tetsuya Shiuchi
- Department of Integrative Physiology, Institute for Health Biosciences, University of Tokushima Graduate School, Tokushima, Japan
| | - Atsushi Kubota
- Department of Sports Medicine, Juntendo University, Chiba, Japan
| | | | - Ryuzo Kawamori
- Department of Metabolism and Endocrinology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Sportology Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Hirotaka Watada
- Department of Metabolism and Endocrinology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Sportology Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Center for Therapeutic Innovations in Diabetes, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Center for Identification of Diabetic Therapeutic Targets, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| |
Collapse
|
18
|
Kanshana JS, Mattila PE, Ewing MC, Wood AN, Schoiswohl G, Meyer AC, Kowalski A, Rosenthal SL, Gingras S, Kaufman BA, Lu R, Weeks DE, McGarvey ST, Minster RL, Hawley NL, Kershaw EE. A murine model of the human CREBRFR457Q obesity-risk variant does not influence energy or glucose homeostasis in response to nutritional stress. PLoS One 2021; 16:e0251895. [PMID: 34520472 PMCID: PMC8439463 DOI: 10.1371/journal.pone.0251895] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/09/2021] [Indexed: 01/02/2023] Open
Abstract
Obesity and diabetes have strong heritable components, yet the genetic contributions to these diseases remain largely unexplained. In humans, a missense variant in Creb3 regulatory factor (CREBRF) [rs373863828 (p.Arg457Gln); CREBRFR457Q] is strongly associated with increased odds of obesity but decreased odds of diabetes. Although virtually nothing is known about CREBRF's mechanism of action, emerging evidence implicates it in the adaptive transcriptional response to nutritional stress downstream of TORC1. The objectives of this study were to generate a murine model with knockin of the orthologous variant in mice (CREBRFR458Q) and to test the hypothesis that this CREBRF variant promotes obesity and protects against diabetes by regulating energy and glucose homeostasis downstream of TORC1. To test this hypothesis, we performed extensive phenotypic analysis of CREBRFR458Q knockin mice at baseline and in response to acute (fasting/refeeding), chronic (low- and high-fat diet feeding), and extreme (prolonged fasting) nutritional stress as well as with pharmacological TORC1 inhibition, and aging to 52 weeks. The results demonstrate that the murine CREBRFR458Q model of the human CREBRFR457Q variant does not influence energy/glucose homeostasis in response to these interventions, with the exception of possible greater loss of fat relative to lean mass with age. Alternative preclinical models and/or studies in humans will be required to decipher the mechanisms linking this variant to human health and disease.
Collapse
Affiliation(s)
- Jitendra S. Kanshana
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Polly E. Mattila
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Michael C. Ewing
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Ashlee N. Wood
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Gabriele Schoiswohl
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria
| | - Anna C. Meyer
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Aneta Kowalski
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Samantha L. Rosenthal
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Sebastien Gingras
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Brett A. Kaufman
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Ray Lu
- Department of Molecular and Cellular Biology, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Daniel E. Weeks
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Stephen T. McGarvey
- International Health Institute, Department of Epidemiology, Brown University School of Public Health, Providence, Rhode Island, United States of America
- Department of Anthropology, Brown University, Providence, Rhode Island, United States of America
| | - Ryan L. Minster
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Nicola L. Hawley
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Erin E. Kershaw
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| |
Collapse
|
19
|
Khromova NV, Fedorov AV, Ma Y, Kondratov KA, Prikhodko SS, Ignatieva EV, Artemyeva MS, Anopova AD, Neimark AE, Kostareva AA, Babenko AY, Dmitrieva RI. Regulatory Action of Plasma from Patients with Obesity and Diabetes towards Muscle Cells Differentiation and Bioenergetics Revealed by the C2C12 Cell Model and MicroRNA Analysis. Biomolecules 2021; 11:769. [PMID: 34063883 PMCID: PMC8224077 DOI: 10.3390/biom11060769] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 11/17/2022] Open
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are often combined and pathologically affect many tissues due to changes in circulating bioactive molecules. In this work, we evaluated the effect of blood plasma from obese (OB) patients or from obese patients comorbid with diabetes (OBD) on skeletal muscle function and metabolic state. We employed the mouse myoblasts C2C12 differentiation model to test the regulatory effect of plasma exposure at several levels: (1) cell morphology; (2) functional activity of mitochondria; (3) expression levels of several mitochondria regulators, i.e., Atgl, Pgc1b, and miR-378a-3p. Existing databases were used to computationally predict and analyze mir-378a-3p potential targets. We show that short-term exposure to OB or OBD patients' plasma is sufficient to affect C2C12 properties. In fact, the expression of genes that regulate skeletal muscle differentiation and growth was downregulated in both OB- and OBD-treated cells, maximal mitochondrial respiration rate was downregulated in the OBD group, while in the OB group, a metabolic switch to glycolysis was detected. These alterations correlated with a decrease in ATGL and Pgc1b expression in the OB group and with an increase of miR-378a-3p levels in the OBD group.
Collapse
Affiliation(s)
- Natalya V. Khromova
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Anton V. Fedorov
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Yi Ma
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Kirill A. Kondratov
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Stanislava S. Prikhodko
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Elena V. Ignatieva
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Marina S. Artemyeva
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Anna D. Anopova
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Aleksandr E. Neimark
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Anna A. Kostareva
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
- Center for Molecular Medicine, Department of Women’s and Children’s Health, Karolinska Institute, 17177 Stockholm, Sweden
| | - Alina Yu. Babenko
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Renata I. Dmitrieva
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| |
Collapse
|
20
|
Turner MC, Rimington RP, Martin NRW, Fleming JW, Capel AJ, Hodson L, Lewis MP. Physiological and pathophysiological concentrations of fatty acids induce lipid droplet accumulation and impair functional performance of tissue engineered skeletal muscle. J Cell Physiol 2021; 236:7033-7044. [PMID: 33738797 DOI: 10.1002/jcp.30365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 02/11/2021] [Accepted: 03/08/2021] [Indexed: 12/19/2022]
Abstract
Fatty acids (FA) exert physiological and pathophysiological effects leading to changes in skeletal muscle metabolism and function, however, in vitro models to investigate these changes are limited. These experiments sought to establish the effects of physiological and pathophysiological concentrations of exogenous FA upon the function of tissue engineered skeletal muscle (TESkM). Cultured initially for 14 days, C2C12 TESkM was exposed to FA-free bovine serum albumin alone or conjugated to a FA mixture (oleic, palmitic, linoleic, and α-linoleic acids [OPLA] [ratio 45:30:24:1%]) at different concentrations (200 or 800 µM) for an additional 4 days. Subsequently, TESkM morphology, functional capacity, gene expression and insulin signaling were analyzed. There was a dose response increase in the number and size of lipid droplets within the TESkM (p < .05). Exposure to exogenous FA increased the messenger RNA expression of genes involved in lipid storage (perilipin 2 [p < .05]) and metabolism (pyruvate dehydrogenase lipoamide kinase isozyme 4 [p < .01]) in a dose dependent manner. TESkM force production was reduced (tetanic and single twitch) (p < .05) and increases in transcription of type I slow twitch fiber isoform, myosin heavy chain 7, were observed when cultured with 200 µM OPLA compared to control (p < .01). Four days of OPLA exposure results in lipid accumulation in TESkM which in turn results in changes in muscle function and metabolism; thus, providing insight ito the functional and mechanistic changes of TESkM in response to exogenous FA.
Collapse
Affiliation(s)
- Mark C Turner
- School of Sport, Exercise and Health Sciences, National Centre for Sport and Exercise Medicine, Loughborough University, Loughborough, UK.,Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust, Leicester, UK.,Centre for Sport, Exercise and Life Sciences, Research Institute for Health and Wellbeing, Coventry University, Coventry, UK
| | - Rowan P Rimington
- School of Sport, Exercise and Health Sciences, National Centre for Sport and Exercise Medicine, Loughborough University, Loughborough, UK
| | - Neil R W Martin
- School of Sport, Exercise and Health Sciences, National Centre for Sport and Exercise Medicine, Loughborough University, Loughborough, UK
| | - Jacob W Fleming
- School of Sport, Exercise and Health Sciences, National Centre for Sport and Exercise Medicine, Loughborough University, Loughborough, UK
| | - Andrew J Capel
- School of Sport, Exercise and Health Sciences, National Centre for Sport and Exercise Medicine, Loughborough University, Loughborough, UK
| | - Leanne Hodson
- Oxford Center for Diabetes, Endocrinology and Metabolism, Oxford Biomedical Research Centre, Radcliffe Department of Medicine, Churchill Hospital, University of Oxford, Oxford, UK
| | - Mark P Lewis
- School of Sport, Exercise and Health Sciences, National Centre for Sport and Exercise Medicine, Loughborough University, Loughborough, UK
| |
Collapse
|
21
|
Kimura K, Morisasa M, Mizushige T, Karasawa R, Kanamaru C, Kabuyama Y, Hayasaka T, Mori T, Goto-Inoue N. Lipid Dynamics due to Muscle Atrophy Induced by Immobilization. J Oleo Sci 2021; 70:937-946. [PMID: 34193670 DOI: 10.5650/jos.ess21045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Muscle atrophy refers to skeletal muscle loss and dysfunction that affects glucose and lipid metabolism. Moreover, muscle atrophy is manifested in cancer, diabetes, and obesity. In this study, we focused on lipid metabolism during muscle atrophy. We observed that the gastrocnemius muscle was associated with significant atrophy with 8 days of immobilization of hind limb joints and that muscle atrophy occurred regardless of the muscle fiber type. Further, we performed lipid analyses using thin layer chromatography, liquid chromatography-mass spectrometry, and mass spectrometry imaging. Total amounts of triacylglycerol, phosphatidylserine, and sphingomyelin were found to be increased in the immobilized muscle. Additionally, we found that specific molecular species of phosphatidylserine, phosphatidylcholine, and sphingomyelin were increased by immobilization. Furthermore, the expression of adipose triglyceride lipase and the activity of cyclooxygenase-2 were significantly reduced by atrophy. From these results, it was revealed that lipid accumulation and metabolic changes in specific fatty acids occur during disuse muscle atrophy. The present study holds implications in validating preventive treatment strategies for muscle atrophy.
Collapse
Affiliation(s)
- Keisuke Kimura
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University
| | - Mizuki Morisasa
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University
| | | | | | | | | | | | - Tsukasa Mori
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University
| | - Naoko Goto-Inoue
- Department of Marine Science and Resources, College of Bioresource Sciences, Nihon University
| |
Collapse
|
22
|
de Oliveira C, Khatua B, Noel P, Kostenko S, Bag A, Balakrishnan B, Patel KS, Guerra AA, Martinez MN, Trivedi S, McCullough A, Lam-Himlin DM, Navina S, Faigel DO, Fukami N, Pannala R, Phillips AE, Papachristou GI, Kershaw EE, Lowe ME, Singh VP. Pancreatic triglyceride lipase mediates lipotoxic systemic inflammation. J Clin Invest 2020; 130:1931-1947. [PMID: 31917686 DOI: 10.1172/jci132767] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 01/03/2020] [Indexed: 12/22/2022] Open
Abstract
Visceral adipose tissue plays a critical role in numerous diseases. Although imaging studies often show adipose involvement in abdominal diseases, their outcomes may vary from being a mild self-limited illness to one with systemic inflammation and organ failure. We therefore compared the pattern of visceral adipose injury during acute pancreatitis and acute diverticulitis to determine its role in organ failure. Acute pancreatitis-associated adipose tissue had ongoing lipolysis in the absence of adipocyte triglyceride lipase (ATGL). Pancreatic lipase injected into mouse visceral adipose tissue hydrolyzed adipose triglyceride and generated excess nonesterified fatty acids (NEFAs), which caused organ failure in the absence of acute pancreatitis. Pancreatic triglyceride lipase (PNLIP) increased in adipose tissue during pancreatitis and entered adipocytes by multiple mechanisms, hydrolyzing adipose triglyceride and generating excess NEFAs. During pancreatitis, obese PNLIP-knockout mice, unlike obese adipocyte-specific ATGL knockouts, had lower visceral adipose tissue lipolysis, milder inflammation, less severe organ failure, and improved survival. PNLIP-knockout mice, unlike ATGL knockouts, were protected from adipocyte-induced pancreatic acinar injury without affecting NEFA signaling or acute pancreatitis induction. Therefore, during pancreatitis, unlike diverticulitis, PNLIP leaking into visceral adipose tissue can cause excessive visceral adipose tissue lipolysis independently of adipocyte-autonomous ATGL, and thereby worsen organ failure.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Ann McCullough
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona, USA
| | - Dora M Lam-Himlin
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona, USA
| | | | | | | | | | - Anna Evans Phillips
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Erin E Kershaw
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mark E Lowe
- Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, USA
| | | |
Collapse
|
23
|
Seibert JT, Najt CP, Heden TD, Mashek DG, Chow LS. Muscle Lipid Droplets: Cellular Signaling to Exercise Physiology and Beyond. Trends Endocrinol Metab 2020; 31:928-938. [PMID: 32917515 PMCID: PMC7704552 DOI: 10.1016/j.tem.2020.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/09/2020] [Accepted: 08/13/2020] [Indexed: 12/21/2022]
Abstract
Conventionally viewed as energy storage depots, lipid droplets (LDs) play a central role in muscle lipid metabolism and intracellular signaling, as recognized by recent advances in our biological understanding. Specific subpopulations of muscle LDs, defined by location and associated proteins, are responsible for distinct biological functions. In this review, the traditional view of muscle LDs is examined, and the emerging role of LDs in intracellular signaling is highlighted. The effects of chronic and acute exercise on muscle LD metabolism and signaling is discussed. In conclusion, future directions for muscle LD research are identified. The primary focus will be on human studies, with inclusion of select animal/cellular/non-muscle studies as appropriate, to provide the underlying mechanisms driving the observed findings.
Collapse
Affiliation(s)
- Jacob T Seibert
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Charles P Najt
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Timothy D Heden
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Douglas G Mashek
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA; Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Minnesota, Minneapolis, MN 55455, USA
| | - Lisa S Chow
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
24
|
Jung YH, Bu SY. Suppression of long chain acyl-CoA synthetase blocks intracellular fatty acid flux and glucose uptake in skeletal myotubes. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158678. [DOI: 10.1016/j.bbalip.2020.158678] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 02/25/2020] [Accepted: 02/28/2020] [Indexed: 12/17/2022]
|
25
|
D’Souza K, Mercer A, Mawhinney H, Pulinilkunnil T, Udenigwe CC, Kienesberger PC. Whey Peptides Stimulate Differentiation and Lipid Metabolism in Adipocytes and Ameliorate Lipotoxicity-Induced Insulin Resistance in Muscle Cells. Nutrients 2020; 12:nu12020425. [PMID: 32041341 PMCID: PMC7071342 DOI: 10.3390/nu12020425] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 01/30/2020] [Accepted: 02/01/2020] [Indexed: 12/13/2022] Open
Abstract
Deregulation of lipid metabolism and insulin function in muscle and adipose tissue are hallmarks of systemic insulin resistance, which can progress to type 2 diabetes. While previous studies suggested that milk proteins influence systemic glucose homeostasis and insulin function, it remains unclear whether bioactive peptides generated from whey alter lipid metabolism and its accumulation in muscle and adipose tissue. Therefore, we incubated murine 3T3-L1 preadipocytes and C2C12 myotubes with a whey peptide mixture produced through pepsin-pancreatin digestion, mimicking peptides generated in the gut from whey protein hydrolysis, and examined its effect on indicators of lipid metabolism and insulin sensitivity. Whey peptides, particularly those derived from bovine serum albumin (BSA), promoted 3T3-L1 adipocyte differentiation and triacylglycerol (TG) accumulation in accordance with peroxisome proliferator-activated receptor γ (PPARγ) upregulation. Whey/BSA peptides also increased lipolysis and mitochondrial fat oxidation in adipocytes, which was associated with the upregulation of peroxisome proliferator-activated receptor δ (PPARδ). In C2C12 myotubes, whey but not BSA peptides ameliorated palmitate-induced insulin resistance, which was associated with reduced inflammation and diacylglycerol accumulation, and increased sequestration of fatty acids in the TG pool. Taken together, our study suggests that whey peptides generated via pepsin-pancreatin digestion profoundly alter lipid metabolism and accumulation in adipocytes and skeletal myotubes.
Collapse
Affiliation(s)
- Kenneth D’Souza
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Dalhousie Medicine, Saint John, NB E2L 4L5 Canada (A.M.); (T.P.)
| | - Angella Mercer
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Dalhousie Medicine, Saint John, NB E2L 4L5 Canada (A.M.); (T.P.)
| | - Hannah Mawhinney
- Department of Plant, Food, and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS B2N 5E3, Canada;
| | - Thomas Pulinilkunnil
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Dalhousie Medicine, Saint John, NB E2L 4L5 Canada (A.M.); (T.P.)
| | - Chibuike C. Udenigwe
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Petra C. Kienesberger
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Dalhousie Medicine, Saint John, NB E2L 4L5 Canada (A.M.); (T.P.)
- Correspondence: ; Tel.: +1-506-636-6971
| |
Collapse
|
26
|
Yu L, Li Y, Grisé A, Wang H. CGI-58: Versatile Regulator of Intracellular Lipid Droplet Homeostasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:197-222. [PMID: 32705602 PMCID: PMC8063591 DOI: 10.1007/978-981-15-6082-8_13] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Comparative gene identification-58 (CGI-58), also known as α/β-hydrolase domain-containing 5 (ABHD5), is a member of a large family of proteins containing an α/β-hydrolase-fold. CGI-58 is well-known as the co-activator of adipose triglyceride lipase (ATGL), which is a key enzyme initiating cytosolic lipid droplet lipolysis. Mutations in either the human CGI-58 or ATGL gene cause an autosomal recessive neutral lipid storage disease, characterized by the excessive accumulation of triglyceride (TAG)-rich lipid droplets in the cytoplasm of almost all cell types. CGI-58, however, has ATGL-independent functions. Distinct phenotypes associated with CGI-58 deficiency commonly include ichthyosis (scaly dry skin), nonalcoholic steatohepatitis, and hepatic fibrosis. Through regulated interactions with multiple protein families, CGI-58 controls many metabolic and signaling pathways, such as lipid and glucose metabolism, energy balance, insulin signaling, inflammatory responses, and thermogenesis. Recent studies have shown that CGI-58 regulates the pathogenesis of common metabolic diseases in a tissue-specific manner. Future studies are needed to molecularly define ATGL-independent functions of CGI-58, including the newly identified serine protease activity of CGI-58. Elucidation of these versatile functions of CGI-58 may uncover fundamental cellular processes governing lipid and energy homeostasis, which may help develop novel approaches that counter against obesity and its associated metabolic sequelae.
Collapse
Affiliation(s)
- Liqing Yu
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Yi Li
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alison Grisé
- College of Computer, Math, and Natural Sciences, College of Behavioral and Social Sciences, University of Maryland, College Park, MD, USA
| | - Huan Wang
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
27
|
Paolella LM, Mukherjee S, Tran CM, Bellaver B, Hugo M, Luongo TS, Shewale SV, Lu W, Chellappa K, Baur JA. mTORC1 restrains adipocyte lipolysis to prevent systemic hyperlipidemia. Mol Metab 2019; 32:136-147. [PMID: 32029223 PMCID: PMC6961719 DOI: 10.1016/j.molmet.2019.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/20/2019] [Accepted: 12/02/2019] [Indexed: 12/11/2022] Open
Abstract
Objective Pharmacological agents targeting the mTOR complexes are used clinically as immunosuppressants and anticancer agents and can extend the lifespan of model organisms. An undesirable side effect of these drugs is hyperlipidemia. Although multiple roles have been described for mTOR complex 1 (mTORC1) in lipid metabolism, the etiology of hyperlipidemia remains incompletely understood. The objective of this study was to determine the influence of adipocyte mTORC1 signaling in systemic lipid homeostasis in vivo. Methods We characterized systemic lipid metabolism in mice lacking the mTORC1 subunit Raptor (RaptoraKO), the key lipolytic enzyme ATGL (ATGLaKO), or both (ATGL-RaptoraKO) in their adipocytes. Results Mice lacking mTORC1 activity in their adipocytes failed to completely suppress lipolysis in the fed state and displayed prominent hypertriglyceridemia and hypercholesterolemia. Blocking lipolysis in their adipose tissue restored normal levels of triglycerides and cholesterol in the fed state as well as the ability to clear triglycerides in an oral fat tolerance test. Conclusions Unsuppressed adipose lipolysis in the fed state interferes with triglyceride clearance and contributes to hyperlipidemia. Adipose tissue mTORC1 activity is necessary for appropriate suppression of lipolysis and for the maintenance of systemic lipid homeostasis. Inhibition of adipose mTORC1 causes hypertriglyceridemia prior to lipodystrophy. Genetically inhibiting lipolysis reverses the increase in plasma TG. Acute pharmacological inhibition of lipolysis reverses the increase in plasma TG caused by rapamycin treatment. Unrestrained lipolysis impairs LPL activity and decreases TG clearance.
Collapse
Affiliation(s)
- Lauren M Paolella
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Institute for Diabetes, Obesity, and Metabolism, Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Sarmistha Mukherjee
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Institute for Diabetes, Obesity, and Metabolism, Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Cassie M Tran
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Institute for Diabetes, Obesity, and Metabolism, Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Bruna Bellaver
- Institute for Diabetes, Obesity, and Metabolism, Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Programa de Pós-graduação em Ciência Biológicas-Bioquímica, Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Mindy Hugo
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Timothy S Luongo
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Institute for Diabetes, Obesity, and Metabolism, Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Swapnil V Shewale
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Wenyun Lu
- Lewis-Sigler Institute for Integrative Genomics, Department of Chemistry, Princeton University, Princeton, NJ, 08544, USA
| | - Karthikeyani Chellappa
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Institute for Diabetes, Obesity, and Metabolism, Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Joseph A Baur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Institute for Diabetes, Obesity, and Metabolism, Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
28
|
Trites MJ, Clugston RD. The role of adipose triglyceride lipase in lipid and glucose homeostasis: lessons from transgenic mice. Lipids Health Dis 2019; 18:204. [PMID: 31757217 PMCID: PMC6874817 DOI: 10.1186/s12944-019-1151-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/07/2019] [Indexed: 12/14/2022] Open
Abstract
The ability of mammals to store and draw on fat reserves has been a driving force throughout evolution in an environment with intermittent nutrient availability. The discovery of adipose triglyceride lipase (ATGL) as a triglyceride lipase provided a heightened understanding of the mechanisms governing mobilization of fat reserves from adipose tissue. ATGL catalyses the initial step in adipose triglyceride lipolysis, working in concert with other enzymes to mobilize triglyceride for energy production. In addition to the role of ATGL in adipose tissue triglyceride mobilization, ATGL plays crucial roles in regulating lipid homeostasis in other tissues. These roles have been characterized primarily using transgenic mice with tissue-specific ATGL ablation. For example, the global ATGL knockout induces a severe cardiac defect that results in premature mortality that is mimicked by inducible cardiomyocyte-specific ATGL knockout. Global- and adipose-specific ATGL ablation induces a whole-body shift from lipid metabolism to glucose metabolism to satisfy metabolic demand primarily facilitated by an increase in glucose uptake by skeletal muscle. Generation of liver-specific ATGL knockouts has implicated hepatic lipolysis as a critical component of normal liver function. Analysis of β-cell ATGL knockouts implicates the necessity of pancreatic ATGL in insulin secretion. The objective of this review is to discuss the contributions of ATGL to systemic lipid- and glucose-homeostasis discovered through the study of transgenic mice.
Collapse
Affiliation(s)
- Michael J Trites
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada.,Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Robin D Clugston
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada. .,Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
29
|
CD8 + T cells induce cachexia during chronic viral infection. Nat Immunol 2019; 20:701-710. [PMID: 31110314 PMCID: PMC6531346 DOI: 10.1038/s41590-019-0397-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/08/2019] [Indexed: 12/19/2022]
Abstract
Cachexia represents a leading cause of morbidity and mortality in various cancers, chronic inflammation and infections. Understanding of the mechanisms that drive cachexia has remained limited, especially for infection-associated cachexia (IAC). Here we describe a model of reversible cachexia in mice with chronic viral infection and identify an essential role for CD8+ T cells in IAC. Cytokines linked to cancer-associated cachexia did not contribute to IAC. Instead, virus-specific CD8+ T cells caused morphological and molecular changes in the adipose tissue, which led to depletion of lipid stores. These changes occurred at a time point that preceded the peak of the CD8+ T cell response and required T cell–intrinsic type 1 interferon signaling and antigen-specific priming. Our results link systemic antiviral immune responses to adipose-tissue remodeling and reveal an underappreciated role of CD8+ T cells in IAC.
Collapse
|
30
|
Supruniuk E, Mikłosz A, Chabowski A, Łukaszuk B. Dose- and time-dependent alterations in lipid metabolism after pharmacological PGC-1α activation in L6 myotubes. J Cell Physiol 2018; 234:11923-11941. [PMID: 30523639 PMCID: PMC6587770 DOI: 10.1002/jcp.27872] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/12/2018] [Indexed: 12/29/2022]
Abstract
Pyrroloquinoline quinone (PQQ) acts as a powerful modulator of PGC‐1α activation and therefore regulates multiple pathways involved in cellular energy homeostasis. In the present study, we assessed the effects of L6 myotubes incubation with 0.5, 1, and 3 μM PQQ solution for 2 and 24 hr with respect to the cells' lipid metabolism. We demonstrated that PQQ significantly elevates PGC‐1α content in a dose‐ and time‐dependent manner with the highest efficiency for 0.5 and 1 µM. The level of free fatty acids was diminished (24 hr: −66%), while an increase in triacylglycerol (TAG) amount was most pronounced after 0.5 μM (2 hr: +93%, 24 hr: +139%) treatment. Ceramide (CER) content was elevated after 2 hr incubation with 0.5 µM and after prolonged exposure to all PQQ concentrations. The cells treated with PQQ for 2 hr exhibited decreased sphinganine (SFA) and sphinganine‐1‐phosphate (SFA1P) level, while 24 hr incubation resulted in an elevated sphingosine (SFO) amount. In summary, PGC‐1α activation promotes TAG and CER synthesis.
Collapse
Affiliation(s)
- Elżbieta Supruniuk
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Agnieszka Mikłosz
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Bartłomiej Łukaszuk
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
31
|
Of mice and men: The physiological role of adipose triglyceride lipase (ATGL). Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1864:880-899. [PMID: 30367950 PMCID: PMC6439276 DOI: 10.1016/j.bbalip.2018.10.008] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/12/2022]
Abstract
Adipose triglyceride lipase (ATGL) has been discovered 14 years ago and revised our view on intracellular triglyceride (TG) mobilization – a process termed lipolysis. ATGL initiates the hydrolysis of TGs to release fatty acids (FAs) that are crucial energy substrates, precursors for the synthesis of membrane lipids, and ligands of nuclear receptors. Thus, ATGL is a key enzyme in whole-body energy homeostasis. In this review, we give an update on how ATGL is regulated on the transcriptional and post-transcriptional level and how this affects the enzymes' activity in the context of neutral lipid catabolism. In depth, we highlight and discuss the numerous physiological functions of ATGL in lipid and energy metabolism. Over more than a decade, different genetic mouse models lacking or overexpressing ATGL in a cell- or tissue-specific manner have been generated and characterized. Moreover, pharmacological studies became available due to the development of a specific murine ATGL inhibitor (Atglistatin®). The identification of patients with mutations in the human gene encoding ATGL and their disease spectrum has underpinned the importance of ATGL in humans. Together, mouse models and human data have advanced our understanding of the physiological role of ATGL in lipid and energy metabolism in adipose and non-adipose tissues, and of the pathophysiological consequences of ATGL dysfunction in mice and men. Summary of mouse models with genetic or pharmacological manipulation of ATGL. Summary of patients with mutations in the human gene encoding ATGL. In depth discussion of the role of ATGL in numerous physiological processes in mice and men.
Collapse
|
32
|
Lee JY, Kim AR, Jung YH, Bu SY. Dissociation of Systemic Glucose Homeostasis from Triacylglyceride Accumulation by Reduced Acsl6 Expression in Skeletal Muscle. BIOTECHNOL BIOPROC E 2018. [DOI: 10.1007/s12257-018-0261-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
33
|
Dai YJ, Liu WB, Li XF, Zhou M, Xu C, Qian Y, Jiang GZ. Molecular cloning of adipose triglyceride lipase (ATGL) gene from blunt snout bream and its expression after LPS-induced TNF-α factor. FISH PHYSIOLOGY AND BIOCHEMISTRY 2018; 44:1143-1157. [PMID: 29705966 DOI: 10.1007/s10695-018-0502-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 04/17/2018] [Indexed: 06/08/2023]
Abstract
The aims of the present study were to clone the full-length cDNA of adipose triglyceridelipase (ATGL) and to analyze its expression after lipopolysaccharide (LPS)-induced tumor necrosis factor alpha (TNF-α). The cDNA obtained covered 1801 bp with an open reading frame of 1500 bp encoding 499 amino acids. Sequence alignment and phylogenetic analysis show the best identity with Cyprinus carpio (86%). The ATGL protein shared a highly conserved 169-amino acid patatin domain, containing a glycine-rich motif, an active serine hydrolase motif, and an aspartic active site. The highest ATGL expression was observed in the liver followed by muscle, whereas relatively low values were detected in the brain and adipose. TNF-α is regarded as an important factor in regulating fat metabolism. Here, LPS was used to induce TNF-α in vivo to verify whether TNF-α can affect ATGL expression. TNF-α expression in liver and muscle is increased and remains unchanged in adipose tissue and brain. The variation of ATGL activity is consistent with that of TNF-α gene expression. Next, we explored the mechanism by which LPS-induced TNF-α mediates the mRNA expression of ATGL in the liver and muscle. For liver, the mRNA levels of c-Jun N-terminal kinase (JNK), nuclear factor kappa B (NF-κB), Sirtuin 1 (SIRT1), and AMP-activated protein kinase (AMPK) were increased by LPS-induced TNF-α. Differencing from the situation in the liver, there was a near-significant decrease trend in the expression of SIRT1 in muscle. Those results indicated that the ATGL gene of blunt snout bream shared a high similarity with the other vertebrates. The expression level of ATGL in tissues with high-fat content was intended to be high. LPS can induce ATGL expression perhaps related to TNF-α.
Collapse
Affiliation(s)
- Yong-Jun Dai
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
| | - Wen-Bin Liu
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
| | - Xiang-Fei Li
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
| | - Man Zhou
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
| | - Chao Xu
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
| | - Yu Qian
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China
| | - Guang-Zhen Jiang
- Key Laboratory of Aquatic Nutrition and Feed Science of Jiangsu Province, College of Animal Science and Technology, Nanjing Agricultural University, No.1 Weigang Road, Nanjing, 210095, People's Republic of China.
| |
Collapse
|
34
|
Schreiber R, Diwoky C, Schoiswohl G, Feiler U, Wongsiriroj N, Abdellatif M, Kolb D, Hoeks J, Kershaw EE, Sedej S, Schrauwen P, Haemmerle G, Zechner R. Cold-Induced Thermogenesis Depends on ATGL-Mediated Lipolysis in Cardiac Muscle, but Not Brown Adipose Tissue. Cell Metab 2017; 26:753-763.e7. [PMID: 28988821 PMCID: PMC5683855 DOI: 10.1016/j.cmet.2017.09.004] [Citation(s) in RCA: 223] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 07/07/2017] [Accepted: 09/05/2017] [Indexed: 11/29/2022]
Abstract
Fatty acids (FAs) activate and fuel UCP1-mediated non-shivering thermogenesis (NST) in brown adipose tissue (BAT). Release of FAs from intracellular fat stores by adipose triglyceride lipase (ATGL) is considered a key step in NST. Accordingly, the severe cold intolerance of global ATGL knockout (AKO) mice has been attributed to defective BAT lipolysis. Here we show that this conclusion is incorrect. We demonstrate that although the BAT-specific loss of ATGL impairs BAT lipolysis and alters BAT morphology, it does not compromise the β3-adrenergic thermogenic response or cold-induced NST. Instead, NST depends on nutrient supply or lipolysis in white adipose tissue during fasting, suggesting that circulating energy substrates are sufficient to fuel NST. Cold intolerance in AKO mice is not caused by BAT dysfunction as previously suspected but by severe cardiomyopathy. We conclude that functional NST requires adequate substrate supply and cardiac function, but does not depend on ATGL-mediated lipolysis in BAT.
Collapse
Affiliation(s)
- Renate Schreiber
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria.
| | - Clemens Diwoky
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | | | - Ursula Feiler
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | | | - Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, 8036 Graz, Austria
| | - Dagmar Kolb
- Institute of Cell Biology, Histology, and Embryology, Core Facility of Ultrastructural Analyses, Medical University of Graz, 8010 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria
| | - Joris Hoeks
- Department of Human Biology and Human Movement Sciences, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Erin E Kershaw
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, 8036 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria
| | - Patrick Schrauwen
- Department of Human Biology and Human Movement Sciences, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Guenter Haemmerle
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria; BioTechMed-Graz, 8010 Graz, Austria.
| |
Collapse
|
35
|
Hua N, Takahashi H, Yee GM, Kitajima Y, Katagiri S, Kojima M, Anzai K, Eguchi Y, Hamilton JA. Influence of muscle fiber type composition on early fat accumulation under high-fat diet challenge. PLoS One 2017; 12:e0182430. [PMID: 28763507 PMCID: PMC5538743 DOI: 10.1371/journal.pone.0182430] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 07/18/2017] [Indexed: 02/03/2023] Open
Abstract
Objective To investigate whether differences in muscle fiber types affect early-stage fat accumulation, under high fat diet challenge in mice. Methods Twelve healthy male C57BL/6 mice experienced with short-term (6 weeks) diet treatment for the evaluation of early pattern changes in muscular fat. The mice were randomly divided into two groups: high fat diet (n = 8) and normal control diet (n = 4). Extra- and intra-myocellular lipid (EMCL and IMCL) in lumbar muscles (type I fiber predominant) and tibialis anterior (TA) muscle (type II fiber predominant) were determined using magnetic resonance spectroscopy (MRS). Correlation of EMCL, IMCL and their ratio between TA and lumbar muscles was evaluated. Results EMCL increased greatly in both muscle types after high fat diet. IMCL in TA and lumbar muscles increased to a much lower extent, with a slightly greater increase in TA muscles. EMCLs in the 2 muscles were positively correlated (r = 0.84, p = 0.01), but IMCLs showed a negative relationship (r = -0.84, p = 0.01). In lumbar muscles, high fat diet significantly decreased type I fiber while it increased type II fiber (all p≤0.001). In TA muscle, there was no significant fiber type shifting (p>0.05). Conclusions Under short-time high fat diet challenge, lipid tends to initially accumulate extra-cellularly. In addition, compared to type II dominant muscle, Type I dominant muscle was less susceptible to IMCL accumulation but more to fiber type shifting. These phenomena might reflect compensative responses of skeletal muscle to dietary lipid overload in order to regulate metabolic homeostasis.
Collapse
Affiliation(s)
- Ning Hua
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA, United States of America
| | - Hirokazu Takahashi
- Research Division, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Internal Medicine, Saga Medical School, Saga, Japan
| | - Grace M. Yee
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA, United States of America
| | - Yoichiro Kitajima
- Division of Internal Medicine, Saga Medical School, Saga, Japan
- Clinical Gastroenterology, Eguchi Hospital, Saga, Japan
| | - Sayaka Katagiri
- Research Division, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Motoyasu Kojima
- Division of Internal Medicine, Saga Medical School, Saga, Japan
| | - Keizo Anzai
- Division of Internal Medicine, Saga Medical School, Saga, Japan
| | - Yuichiro Eguchi
- Division of Hepatology, Saga Medical School, Liver Center, Saga, Japan
| | - James A. Hamilton
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA, United States of America
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
36
|
Scerbo D, Son NH, Sirwi A, Zeng L, Sas KM, Cifarelli V, Schoiswohl G, Huggins LA, Gumaste N, Hu Y, Pennathur S, Abumrad NA, Kershaw EE, Hussain MM, Susztak K, Goldberg IJ. Kidney triglyceride accumulation in the fasted mouse is dependent upon serum free fatty acids. J Lipid Res 2017; 58:1132-1142. [PMID: 28404638 DOI: 10.1194/jlr.m074427] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 04/10/2017] [Indexed: 01/13/2023] Open
Abstract
Lipid accumulation is a pathological feature of every type of kidney injury. Despite this striking histological feature, physiological accumulation of lipids in the kidney is poorly understood. We studied whether the accumulation of lipids in the fasted kidney are derived from lipoproteins or NEFAs. With overnight fasting, kidneys accumulated triglyceride, but had reduced levels of ceramide and glycosphingolipid species. Fasting led to a nearly 5-fold increase in kidney uptake of plasma [14C]oleic acid. Increasing circulating NEFAs using a β adrenergic receptor agonist caused a 15-fold greater accumulation of lipid in the kidney, while mice with reduced NEFAs due to adipose tissue deficiency of adipose triglyceride lipase had reduced triglycerides. Cluster of differentiation (Cd)36 mRNA increased 2-fold, and angiopoietin-like 4 (Angptl4), an LPL inhibitor, increased 10-fold. Fasting-induced kidney lipid accumulation was not affected by inhibition of LPL with poloxamer 407 or by use of mice with induced genetic LPL deletion. Despite the increase in CD36 expression with fasting, genetic loss of CD36 did not alter fatty acid uptake or triglyceride accumulation. Our data demonstrate that fasting-induced triglyceride accumulation in the kidney correlates with the plasma concentrations of NEFAs, but is not due to uptake of lipoprotein lipids and does not involve the fatty acid transporter, CD36.
Collapse
Affiliation(s)
- Diego Scerbo
- Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York, NY.,Institute of Human Nutrition, Columbia University, New York, NY
| | - Ni-Huiping Son
- Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York, NY
| | - Alaa Sirwi
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY
| | - Lixia Zeng
- Division of Nephrology, University of Michigan, Ann Arbor, MI
| | - Kelli M Sas
- Division of Nephrology, University of Michigan, Ann Arbor, MI
| | | | - Gabriele Schoiswohl
- Division of Endocrinology, University of Pittsburgh, Pittsburgh, PA.,Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Lesley-Ann Huggins
- Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York, NY
| | - Namrata Gumaste
- Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York, NY
| | - Yunying Hu
- Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York, NY
| | | | - Nada A Abumrad
- Department of Medicine, Washington University, St. Louis, MO
| | - Erin E Kershaw
- Division of Endocrinology, University of Pittsburgh, Pittsburgh, PA
| | - M Mahmood Hussain
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY
| | - Katalin Susztak
- Division of Renal Electrolyte and Hypertension, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ira J Goldberg
- Division of Endocrinology, Diabetes, and Metabolism, New York University School of Medicine, New York, NY
| |
Collapse
|
37
|
Laurens C, Moro C. Intramyocellular fat storage in metabolic diseases. Horm Mol Biol Clin Investig 2017; 26:43-52. [PMID: 26741351 DOI: 10.1515/hmbci-2015-0045] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 11/18/2015] [Indexed: 12/13/2022]
Abstract
Over the past decades, obesity and its metabolic co-morbidities such as type 2 diabetes (T2D) developed to reach an endemic scale. However, the mechanisms leading to the development of T2D are still poorly understood. One main predictor for T2D seems to be lipid accumulation in "non-adipose" tissues, best known as ectopic lipid storage. A growing body of data suggests that these lipids may play a role in impairing insulin action in metabolic tissues, such as liver and skeletal muscle. This review aims to discuss recent literature linking ectopic lipid storage and insulin resistance, with emphasis on lipid deposition in skeletal muscle. The link between skeletal muscle lipid content and insulin sensitivity, as well as the mechanisms of lipid-induced insulin resistance and potential therapeutic strategies to alleviate lipotoxic lipid pressure in skeletal muscle will be discussed.
Collapse
|
38
|
Wang L, Lin G, Lee YC, Reed-Maldonado AB, Sanford MT, Wang G, Li H, Banie L, Xin Z, Lue TF. Transgenic animal model for studying the mechanism of obesity-associated stress urinary incontinence. BJU Int 2016; 119:317-324. [PMID: 27649937 DOI: 10.1111/bju.13661] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVE To study and compare the function and structure of the urethral sphincter in female Zucker lean (ZL) and Zucker fatty (ZF) rats and to assess the viability of ZF fats as a model for female obesity-associated stress urinary incontinence (SUI). MATERIALS AND METHODS Two study arms were created: a ZL arm including 16-week-old female ZL rats (ZUC-Leprfa 186; n = 12) and a ZF arm including 16-week-old female ZF rats (ZUC-Leprfa 185; n = 12). I.p. insulin tolerance testing was carried out before functional study. Metabolic cages, conscious cystometry and leak point pressure (LPP) assessments were conducted. Urethral tissues were harvested for immunofluorescence staining to check intramyocellular lipid (IMCL) and sphincter muscle (smooth muscle and striated muscle) composition. RESULTS The ZF rats had insulin resistance, a greater voiding frequency and lower LPP compared with ZL rats (P < 0.05), with more IMCL deposition localized in the urethral striated muscle fibres of the ZF rats (P < 0.05). The thickness of the striated muscle layer and the ratio of striated muscle to smooth muscle were lower in ZF than in ZL rats. CONCLUSION Obesity impairs urethral sphincter function via IMCL deposition and leads to atrophy and distortion of urethral striated muscle. The ZF rats could be a consistent and reliable animal model in which to study obesity-associated SUI.
Collapse
Affiliation(s)
- Lin Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA.,Department of Urology, Peking University First Hospital, Peking University, Beijing, China
| | - Guiting Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Yung-Chin Lee
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA.,Department of Urology, Faculty of Medicine, Kaohsiung Medical University Hospital, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Amanda B Reed-Maldonado
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Melissa T Sanford
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Guifang Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Huixi Li
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA.,Department of Urology, Peking University First Hospital, Peking University, Beijing, China
| | - Lia Banie
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Zhengcheng Xin
- Department of Urology, Peking University First Hospital, Peking University, Beijing, China
| | - Tom F Lue
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
39
|
Dirks ML, Wall BT, van de Valk B, Holloway TM, Holloway GP, Chabowski A, Goossens GH, van Loon LJC. One Week of Bed Rest Leads to Substantial Muscle Atrophy and Induces Whole-Body Insulin Resistance in the Absence of Skeletal Muscle Lipid Accumulation. Diabetes 2016; 65:2862-75. [PMID: 27358494 DOI: 10.2337/db15-1661] [Citation(s) in RCA: 266] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 06/23/2016] [Indexed: 11/13/2022]
Abstract
Short (<10 days) periods of muscle disuse, often necessary for recovery from illness or injury, lead to various negative health consequences. The current study investigated mechanisms underlying disuse-induced insulin resistance, taking into account muscle atrophy. Ten healthy, young males (age: 23 ± 1 years; BMI: 23.0 ± 0.9 kg · m(-2)) were subjected to 1 week of strict bed rest. Prior to and after bed rest, lean body mass (dual-energy X-ray absorptiometry) and quadriceps cross-sectional area (CSA; computed tomography) were assessed, and peak oxygen uptake (VO2peak) and leg strength were determined. Whole-body insulin sensitivity was measured using a hyperinsulinemic-euglycemic clamp. Additionally, muscle biopsies were collected to assess muscle lipid (fraction) content and various markers of mitochondrial and vascular content. Bed rest resulted in 1.4 ± 0.2 kg lean tissue loss and a 3.2 ± 0.9% decline in quadriceps CSA (both P < 0.01). VO2peak and one-repetition maximum declined by 6.4 ± 2.3 (P < 0.05) and 6.9 ± 1.4% (P < 0.01), respectively. Bed rest induced a 29 ± 5% decrease in whole-body insulin sensitivity (P < 0.01). This was accompanied by a decline in muscle oxidative capacity, without alterations in skeletal muscle lipid content or saturation level, markers of oxidative stress, or capillary density. In conclusion, 1 week of bed rest substantially reduces skeletal muscle mass and lowers whole-body insulin sensitivity, without affecting mechanisms implicated in high-fat diet-induced insulin resistance.
Collapse
Affiliation(s)
- Marlou L Dirks
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastrict, the Netherlands
| | - Benjamin T Wall
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastrict, the Netherlands
| | - Bas van de Valk
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastrict, the Netherlands
| | - Tanya M Holloway
- Human Health & Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Graham P Holloway
- Human Health & Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Gijs H Goossens
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastrict, the Netherlands
| | - Luc J C van Loon
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastrict, the Netherlands
| |
Collapse
|
40
|
Biswas D, Ghosh M, Kumar S, Chakrabarti P. PPARα-ATGL pathway improves muscle mitochondrial metabolism: implication in aging. FASEB J 2016; 30:3822-3834. [PMID: 27485820 DOI: 10.1096/fj.201600571rr] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 07/27/2016] [Indexed: 02/05/2023]
Abstract
Adipose triglyceride lipase (ATGL) maintains an optimum mitochondrial function putatively by generating cognate ligands for peroxisome proliferator-activated receptor α (PPARα), which, together with PPARγ coactivator-1α (PGC1α), regulate muscle mitochondrial biogenesis. However, the cross-talk between ATGL and PPARα in skeletal muscle mitochondrial metabolism and its implication in chronological aging is poorly understood. The role of ATGL in muscle mitochondrial metabolism was studied by overexpressing and depleting the gene and studying its downstream effect in cultured myotubes and in murine skeletal muscle. We found that PPARα directly induces ATGL expression during myogenesis. Overexpression of ATGL significantly enhanced while depletion of ATGL attenuated mitochondrial oxidative phosphorylation and fatty acid oxidation without alteration in mitochondrial content, and it rendered PPARα and PGC1α redundant in promoting mitochondrial oxidative function. However, ATGL did not alter PPARα-dependent lipid accumulation and insulin sensitivity. In middle-aged rats, ATGL expression was higher and correlated with PPARα expression and sustained fatty acid oxidation in oxidative soleus muscle. Fenofibrate feeding further induced ATGL expression selectively in this muscle compartment. These findings illustrate that PPARα and ATGL constitute a regulatory pathway in skeletal muscle, suggesting their role as a mitochondrial metabolic reserve.-Biswas, D., Ghosh, M., Kumar, S., Chakrabarti, P. PPARα-ATGL pathway improves muscle mitochondrial metabolism: implication in aging.
Collapse
Affiliation(s)
- Dipsikha Biswas
- Division of Cell Biology and Physiology, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India; and
| | - Mainak Ghosh
- Division of Cell Biology and Physiology, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India; and
| | - Subhankar Kumar
- Department of Physiology, Institute of Postgraduate Medical Education and Research, Kolkata, India
| | - Partha Chakrabarti
- Division of Cell Biology and Physiology, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India; and
| |
Collapse
|
41
|
Sun J, Ji H, Li XX, Shi XC, Du ZY, Chen LQ. Lipolytic enzymes involving lipolysis in Teleost: Synteny, structure, tissue distribution, and expression in grass carp (Ctenopharyngodon idella). Comp Biochem Physiol B Biochem Mol Biol 2016; 198:110-8. [DOI: 10.1016/j.cbpb.2016.04.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 04/25/2016] [Accepted: 04/25/2016] [Indexed: 02/06/2023]
|
42
|
Lord CC, Ferguson D, Thomas G, Brown AL, Schugar RC, Burrows A, Gromovsky AD, Betters J, Neumann C, Sacks J, Marshall S, Watts R, Schweiger M, Lee RG, Crooke RM, Graham MJ, Lathia JD, Sakaguchi TF, Lehner R, Haemmerle G, Zechner R, Brown JM. Regulation of Hepatic Triacylglycerol Metabolism by CGI-58 Does Not Require ATGL Co-activation. Cell Rep 2016; 16:939-949. [PMID: 27396333 DOI: 10.1016/j.celrep.2016.06.049] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/20/2016] [Accepted: 06/10/2016] [Indexed: 01/23/2023] Open
Abstract
Adipose triglyceride lipase (ATGL) and comparative gene identification 58 (CGI-58) are critical regulators of triacylglycerol (TAG) turnover. CGI-58 is thought to regulate TAG mobilization by stimulating the enzymatic activity of ATGL. However, it is not known whether this coactivation function of CGI-58 occurs in vivo. Moreover, the phenotype of human CGI-58 mutations suggests ATGL-independent functions. Through direct comparison of mice with single or double deficiency of CGI-58 and ATGL, we show here that CGI-58 knockdown causes hepatic steatosis in both the presence and absence of ATGL. CGI-58 also regulates hepatic diacylglycerol (DAG) and inflammation in an ATGL-independent manner. Interestingly, ATGL deficiency, but not CGI-58 deficiency, results in suppression of the hepatic and adipose de novo lipogenic program. Collectively, these findings show that CGI-58 regulates hepatic neutral lipid storage and inflammation in the genetic absence of ATGL, demonstrating that mechanisms driving TAG lipolysis in hepatocytes differ significantly from those in adipocytes.
Collapse
Affiliation(s)
- Caleb C Lord
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1040, USA; Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9077, USA
| | - Daniel Ferguson
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1040, USA; Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Gwynneth Thomas
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1040, USA
| | - Amanda L Brown
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1040, USA; Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Rebecca C Schugar
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Amy Burrows
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Anthony D Gromovsky
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jenna Betters
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1040, USA
| | - Chase Neumann
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jessica Sacks
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Stephanie Marshall
- Section on Lipid Sciences, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC 27157-1040, USA; Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Russell Watts
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Martina Schweiger
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Richard G Lee
- Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | - Rosanne M Crooke
- Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | - Mark J Graham
- Cardiovascular Group, Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | - Justin D Lathia
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Takuya F Sakaguchi
- Department of Stem Cell Biology and Regenerative Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Richard Lehner
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Guenter Haemmerle
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - J Mark Brown
- Department of Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
43
|
Schoiswohl G, Stefanovic-Racic M, Menke MN, Wills RC, Surlow BA, Basantani MK, Sitnick MT, Cai L, Yazbeck CF, Stolz DB, Pulinilkunnil T, O'Doherty RM, Kershaw EE. Impact of Reduced ATGL-Mediated Adipocyte Lipolysis on Obesity-Associated Insulin Resistance and Inflammation in Male Mice. Endocrinology 2015; 156. [PMID: 26196542 PMCID: PMC4588821 DOI: 10.1210/en.2015-1322] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Emerging evidence suggests that impaired regulation of adipocyte lipolysis contributes to the proinflammatory immune cell infiltration of metabolic tissues in obesity, a process that is proposed to contribute to the development and exacerbation of insulin resistance. To test this hypothesis in vivo, we generated mice with adipocyte-specific deletion of adipose triglyceride lipase (ATGL), the rate-limiting enzyme catalyzing triacylglycerol hydrolysis. In contrast to previous models, adiponectin-driven Cre expression was used for targeted ATGL deletion. The resulting adipocyte-specific ATGL knockout (AAKO) mice were then characterized for metabolic and immune phenotypes. Lean and diet-induced obese AAKO mice had reduced adipocyte lipolysis, serum lipids, systemic lipid oxidation, and expression of peroxisome proliferator-activated receptor alpha target genes in adipose tissue (AT) and liver. These changes did not increase overall body weight or fat mass in AAKO mice by 24 weeks of age, in part due to reduced expression of genes involved in lipid uptake, synthesis, and adipogenesis. Systemic glucose and insulin tolerance were improved in AAKO mice, primarily due to enhanced hepatic insulin signaling, which was accompanied by marked reduction in diet-induced hepatic steatosis as well as hepatic immune cell infiltration and activation. In contrast, although adipocyte ATGL deletion reduced AT immune cell infiltration in response to an acute lipolytic stimulus, it was not sufficient to ameliorate, and may even exacerbate, chronic inflammatory changes that occur in AT in response to diet-induced obesity.
Collapse
Affiliation(s)
- Gabriele Schoiswohl
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Maja Stefanovic-Racic
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Marie N Menke
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Rachel C Wills
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Beth A Surlow
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Mahesh K Basantani
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Mitch T Sitnick
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Lingzhi Cai
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Cynthia F Yazbeck
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Donna B Stolz
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Thomas Pulinilkunnil
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Robert M O'Doherty
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| | - Erin E Kershaw
- Division of Endocrinology and Metabolism (G.S., M.S.-R., R.C.W., B.A.S., M.K.B., M.T.S., L.C., C.F.Y., R.M.O., E.E.K.), Department of Medicine, and Department of Cell Biology (D.B.S.), University of Pittsburgh, Pittsburgh, Pennsylvania 15261; Department of Obstetrics, Gynecology, and Reproductive Sciences (M.N.M.), Magee-Womens Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania 15213; and Department of Biochemistry and Molecular Biology (T.P.), Dalhousie Medicine New Brunswick, Dalhousie University, St John, Canada NB E2L 4L5
| |
Collapse
|
44
|
Bonen A, Jain SS, Snook LA, Han XX, Yoshida Y, Buddo KH, Lally JS, Pask ED, Paglialunga S, Beaudoin MS, Glatz JFC, Luiken JJFP, Harasim E, Wright DC, Chabowski A, Holloway GP. Extremely rapid increase in fatty acid transport and intramyocellular lipid accumulation but markedly delayed insulin resistance after high fat feeding in rats. Diabetologia 2015. [PMID: 26197708 DOI: 10.1007/s00125-015-3691-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS The mechanisms for diet-induced intramyocellular lipid accumulation and its association with insulin resistance remain contentious. In a detailed time-course study in rats, we examined whether a high-fat diet increased intramyocellular lipid accumulation via alterations in fatty acid translocase (FAT/CD36)-mediated fatty acid transport, selected enzymes and/or fatty acid oxidation, and whether intramyocellular lipid accretion coincided with the onset of insulin resistance. METHODS We measured, daily (on days 1-7) and/or weekly (for 6 weeks), the diet-induced changes in circulating substrates, insulin, sarcolemmal substrate transporters and transport, selected enzymes, intramyocellular lipids, mitochondrial fatty acid oxidation and basal and insulin-stimulated sarcolemmal GLUT4 and glucose transport. We also examined whether upregulating fatty acid oxidation improved glucose transport in insulin-resistant muscles. Finally, in Cd36-knockout mice, we examined the role of FAT/CD36 in intramyocellular lipid accumulation, insulin sensitivity and diet-induced glucose intolerance. RESULTS Within 2-3 days, diet-induced increases occurred in insulin, sarcolemmal FAT/CD36 (but not fatty acid binding protein [FABPpm] or fatty acid transporter [FATP]1 or 4), fatty acid transport and intramyocellular triacylglycerol, diacylglycerol and ceramide, independent of enzymatic changes or muscle fatty acid oxidation. Diet-induced increases in mitochondria and mitochondrial fatty acid oxidation and impairments in insulin-stimulated glucose transport and GLUT4 translocation occurred much later (≥21 days). FAT/CD36 ablation impaired insulin-stimulated fatty acid transport and lipid accumulation, improved insulin sensitivity and prevented diet-induced glucose intolerance. Increasing fatty acid oxidation in insulin-resistant muscles improved glucose transport. CONCLUSIONS/INTERPRETATIONS High-fat feeding rapidly increases intramyocellular lipids (in 2-3 days) via insulin-mediated upregulation of sarcolemmal FAT/CD36 and fatty acid transport. The 16-19 day delay in the onset of insulin resistance suggests that additional mechanisms besides intramyocellular lipids contribute to this pathology.
Collapse
Affiliation(s)
- Arend Bonen
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1.
| | - Swati S Jain
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - Laelie A Snook
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - Xiao-Xia Han
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - Yuko Yoshida
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - Kathryn H Buddo
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - James S Lally
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - Elizabeth D Pask
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - Sabina Paglialunga
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - Marie-Soleil Beaudoin
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - Jan F C Glatz
- Department of Molecular Genetics, Maastricht University, Maastricht, the Netherlands
| | - Joost J F P Luiken
- Department of Molecular Genetics, Maastricht University, Maastricht, the Netherlands
| | - Ewa Harasim
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - David C Wright
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Graham P Holloway
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada, N1G 2W1
| |
Collapse
|
45
|
Foryst-Ludwig A, Kreissl MC, Benz V, Brix S, Smeir E, Ban Z, Januszewicz E, Salatzki J, Grune J, Schwanstecher AK, Blumrich A, Schirbel A, Klopfleisch R, Rothe M, Blume K, Halle M, Wolfarth B, Kershaw EE, Kintscher U. Adipose Tissue Lipolysis Promotes Exercise-induced Cardiac Hypertrophy Involving the Lipokine C16:1n7-Palmitoleate. J Biol Chem 2015; 290:23603-15. [PMID: 26260790 DOI: 10.1074/jbc.m115.645341] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Indexed: 12/28/2022] Open
Abstract
Endurance exercise training induces substantial adaptive cardiac modifications such as left ventricular hypertrophy (LVH). Simultaneously to the development of LVH, adipose tissue (AT) lipolysis becomes elevated upon endurance training to cope with enhanced energy demands. In this study, we investigated the impact of adipose tissue lipolysis on the development of exercise-induced cardiac hypertrophy. Mice deficient for adipose triglyceride lipase (Atgl) in AT (atATGL-KO) were challenged with chronic treadmill running. Exercise-induced AT lipolytic activity was significantly reduced in atATGL-KO mice accompanied by the absence of a plasma fatty acid (FA) increase. These processes were directly associated with a prominent attenuation of myocardial FA uptake in atATGL-KO and a significant reduction of the cardiac hypertrophic response to exercise. FA serum profiling revealed palmitoleic acid (C16:1n7) as a new molecular co-mediator of exercise-induced cardiac hypertrophy by inducing nonproliferative cardiomyocyte growth. In parallel, serum FA analysis and echocardiography were performed in 25 endurance athletes. In consonance, the serum C16:1n7 palmitoleate level exhibited a significantly positive correlation with diastolic interventricular septum thickness in those athletes. No correlation existed between linoleic acid (18:2n6) and diastolic interventricular septum thickness. Collectively, our data provide the first evidence that adipose tissue lipolysis directly promotes the development of exercise-induced cardiac hypertrophy involving the lipokine C16:1n7 palmitoleate as a molecular co-mediator. The identification of a lipokine involved in physiological cardiac growth may help to develop future lipid-based therapies for pathological LVH or heart failure.
Collapse
Affiliation(s)
- Anna Foryst-Ludwig
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany, the DZHK (German Center for Cardiovascular Research), 10115 Berlin, Germany
| | - Michael C Kreissl
- the Department of Nuclear Medicine, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
| | - Verena Benz
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Sarah Brix
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Elia Smeir
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Zsofia Ban
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Elżbieta Januszewicz
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Janek Salatzki
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Jana Grune
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Anne-Kathrin Schwanstecher
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Annelie Blumrich
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Andreas Schirbel
- the Department of Nuclear Medicine, University Hospital Wuerzburg, 97080 Wuerzburg, Germany
| | - Robert Klopfleisch
- the Department of Veterinary Pathology, College of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany
| | | | - Katharina Blume
- the Department of Prevention, Rehabilitation, and Sports Medicine, Technische Universitaet Muenchen, 80809 Muenchen, Germany
| | - Martin Halle
- the Department of Prevention, Rehabilitation, and Sports Medicine, Technische Universitaet Muenchen, 80809 Muenchen, Germany, the DZHK (German Center for Cardiovascular Research), Munich Heart Alliance, 80809 Munich, Germany
| | - Bernd Wolfarth
- the Department of Sports Medicine, Humboldt University/Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany
| | - Erin E Kershaw
- the Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, and
| | - Ulrich Kintscher
- From the Institute of Pharmacology, Center for Cardiovascular Research, Charité-Universitaetsmedizin Berlin, 10115 Berlin, Germany, the DZHK (German Center for Cardiovascular Research), 10115 Berlin, Germany
| |
Collapse
|
46
|
Jaeger D, Schoiswohl G, Hofer P, Schreiber R, Schweiger M, Eichmann TO, Pollak NM, Poecher N, Grabner GF, Zierler KA, Eder S, Kolb D, Radner FPW, Preiss-Landl K, Lass A, Zechner R, Kershaw EE, Haemmerle G. Fasting-induced G0/G1 switch gene 2 and FGF21 expression in the liver are under regulation of adipose tissue derived fatty acids. J Hepatol 2015; 63:437-45. [PMID: 25733154 PMCID: PMC4518503 DOI: 10.1016/j.jhep.2015.02.035] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 02/18/2015] [Accepted: 02/20/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Adipose tissue (AT)-derived fatty acids (FAs) are utilized for hepatic triacylglycerol (TG) generation upon fasting. However, their potential impact as signaling molecules is not established. Herein we examined the role of exogenous AT-derived FAs in the regulation of hepatic gene expression by investigating mice with a defect in AT-derived FA supply to the liver. METHODS Plasma FA levels, tissue TG hydrolytic activities and lipid content were determined in mice lacking the lipase co-activator comparative gene identification-58 (CGI-58) selectively in AT (CGI-58-ATko) applying standard protocols. Hepatic expression of lipases, FA oxidative genes, transcription factors, ER stress markers, hormones and cytokines were determined by qRT-PCR, Western blotting and ELISA. RESULTS Impaired AT-derived FA supply upon fasting of CGI-58-ATko mice causes a marked defect in liver PPARα-signaling and nuclear CREBH translocation. This severely reduced the expression of respective target genes such as the ATGL inhibitor G0/G1 switch gene-2 (G0S2) and the endocrine metabolic regulator FGF21. These changes could be reversed by lipid administration and raising plasma FA levels. Impaired AT-lipolysis failed to induce hepatic G0S2 expression in fasted CGI-58-ATko mice leading to enhanced ATGL-mediated TG-breakdown strongly reducing hepatic TG deposition. On high fat diet, impaired AT-lipolysis counteracts hepatic TG accumulation and liver stress linked to improved systemic insulin sensitivity. CONCLUSIONS AT-derived FAs are a critical regulator of hepatic fasting gene expression required for the induction of G0S2-expression in the liver to control hepatic TG-breakdown. Interfering with AT-lipolysis or hepatic G0S2 expression represents an effective strategy for the treatment of hepatic steatosis.
Collapse
Affiliation(s)
- Doris Jaeger
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Gabriele Schoiswohl
- Division of Endocrinology, Diabetes, and Metabolism, University of Pittsburgh, PA 15261, USA
| | - Peter Hofer
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Renate Schreiber
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Martina Schweiger
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Thomas O Eichmann
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Nina M Pollak
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Nadja Poecher
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Gernot F Grabner
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Kathrin A Zierler
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Sandra Eder
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Dagmar Kolb
- ZMF, Center for Medical Research, Medical University of Graz, A-8010 Graz, Austria
| | - Franz P W Radner
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Karina Preiss-Landl
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Achim Lass
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria
| | - Erin E Kershaw
- Division of Endocrinology, Diabetes, and Metabolism, University of Pittsburgh, PA 15261, USA
| | - Guenter Haemmerle
- Institute of Molecular Biosciences, University of Graz, Heinrichstrasse 31, A-8010 Graz, Austria.
| |
Collapse
|
47
|
Meex RCR, Hoy AJ, Mason RM, Martin SD, McGee SL, Bruce CR, Watt MJ. ATGL-mediated triglyceride turnover and the regulation of mitochondrial capacity in skeletal muscle. Am J Physiol Endocrinol Metab 2015; 308:E960-70. [PMID: 25852007 DOI: 10.1152/ajpendo.00598.2014] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 04/02/2015] [Indexed: 12/11/2022]
Abstract
Emerging evidence indicates that skeletal muscle lipid droplets are an important control point for intracellular lipid homeostasis and that regulating fatty acid fluxes from lipid droplets might influence mitochondrial capacity. We used pharmacological blockers of the major triglyceride lipases, adipose triglyceride lipase (ATGL) and hormone-sensitive lipase, to show that a large proportion of the fatty acids that are transported into myotubes are trafficked through the intramyocellular triglyceride pool. We next tested whether increasing lipolysis from intramyocellular lipid droplets could activate transcriptional responses to enhance mitochondrial and fatty acid oxidative capacity. ATGL was overexpressed by adenoviral and adenoassociated viral infection in C2C12 myotubes and the tibialis anterior muscle of C57Bl/6 mice, respectively. ATGL overexpression in C2C12 myotubes increased lipolysis, which was associated with increased peroxisome proliferator-activated receptor (PPAR)-∂ activity, transcriptional upregulation of some PPAR∂ target genes, and enhanced mitochondrial capacity. The transcriptional responses were specific to ATGL actions and not a generalized increase in fatty acid flux in the myotubes. Marked ATGL overexpression (20-fold) induced modest molecular changes in the skeletal muscle of mice, but these effects were not sufficient to alter fatty acid oxidation. Together, these data demonstrate the importance of lipid droplets for myocellular fatty acid trafficking and the capacity to modulate mitochondrial capacity by enhancing lipid droplet lipolysis in vitro; however, this adaptive program is of minor importance when superimposing the normal metabolic stresses encountered in free-moving animals.
Collapse
Affiliation(s)
- Ruth C R Meex
- Biology of Lipid Metabolism Laboratory, Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Andrew J Hoy
- Discipline of Physiology, School of Medical Sciences & Bosch Institute, University of Sydney, New South Wales, Australia; Boden Institute of Obesity, Nutrition, Exercise & Eating Disorders, University of Sydney, Sydney, New South Wales, Australia
| | - Rachael M Mason
- Biology of Lipid Metabolism Laboratory, Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Sheree D Martin
- Metabolic Remodelling Laboratory, Metabolic Research Unit, School of Medicine, Deakin University, Burwood, Victoria, Australia; and
| | - Sean L McGee
- Metabolic Remodelling Laboratory, Metabolic Research Unit, School of Medicine, Deakin University, Burwood, Victoria, Australia; and
| | - Clinton R Bruce
- Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Victoria, Australia
| | - Matthew J Watt
- Biology of Lipid Metabolism Laboratory, Department of Physiology, Monash University, Clayton, Victoria, Australia;
| |
Collapse
|
48
|
Dubé JJ, Sitnick MT, Schoiswohl G, Wills RC, Basantani MK, Cai L, Pulinilkunnil T, Kershaw EE. Adipose triglyceride lipase deletion from adipocytes, but not skeletal myocytes, impairs acute exercise performance in mice. Am J Physiol Endocrinol Metab 2015; 308:E879-90. [PMID: 25783895 PMCID: PMC4436997 DOI: 10.1152/ajpendo.00530.2014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 03/09/2015] [Indexed: 12/22/2022]
Abstract
Adipose triglyceride lipase (ATGL) is the rate-limiting enzyme mediating triacylglycerol hydrolysis in virtually all cells, including adipocytes and skeletal myocytes, and hence, plays a critical role in mobilizing fatty acids. Global ATGL deficiency promotes skeletal myopathy and exercise intolerance in mice and humans, and yet the tissue-specific contributions to these phenotypes remain unknown. The goal of this study was to determine the relative contribution of ATGL-mediated triacylglycerol hydrolysis in adipocytes vs. skeletal myocytes to acute exercise performance. To achieve this goal, we generated murine models with adipocyte- and skeletal myocyte-specific targeted deletion of ATGL. We then subjected untrained mice to acute peak and submaximal exercise interventions and assessed exercise performance and energy substrate metabolism. Impaired ATGL-mediated lipolysis within adipocytes reduced peak and submaximal exercise performance, reduced peripheral energy substrate availability, shifted energy substrate preference toward carbohydrate oxidation, and decreased HSL Ser(660) phosphorylation and mitochondrial respiration within skeletal muscle. In contrast, impaired ATGL-mediated lipolysis within skeletal myocytes was not sufficient to reduce peak and submaximal exercise performance or peripheral energy substrate availability and instead tended to enhance metabolic flexibility during peak exercise. Furthermore, the expanded intramyocellular triacylglycerol pool in these mice was reduced following exercise in association with preserved HSL phosphorylation, suggesting that HSL may compensate for impaired ATGL action in skeletal muscle during exercise. These data suggest that adipocyte rather than skeletal myocyte ATGL-mediated lipolysis plays a greater role during acute exercise in part because of compensatory mechanisms that maintain lipolysis in muscle, but not adipose tissue, when ATGL is absent.
Collapse
Affiliation(s)
- John J Dubé
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Mitch T Sitnick
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Gabriele Schoiswohl
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Rachel C Wills
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Mahesh K Basantani
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Lingzhi Cai
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Thomas Pulinilkunnil
- Department of Biochemistry and Molecular Biology, Dalhousie Medicine New Brunswick, Dalhousie University, Saint John, New Brunswick, Canada
| | - Erin E Kershaw
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| |
Collapse
|
49
|
Xie P, Kadegowda AKG, Ma Y, Guo F, Han X, Wang M, Groban L, Xue B, Shi H, Li H, Yu L. Muscle-specific deletion of comparative gene identification-58 (CGI-58) causes muscle steatosis but improves insulin sensitivity in male mice. Endocrinology 2015; 156:1648-58. [PMID: 25751639 PMCID: PMC4398773 DOI: 10.1210/en.2014-1892] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Intramyocellular accumulation of lipids is often associated with insulin resistance. Deficiency of comparative gene identification-58 (CGI-58) causes cytosolic deposition of triglyceride (TG)-rich lipid droplets in most cell types, including muscle due to defective TG hydrolysis. It was unclear, however, whether CGI-58 deficiency-induced lipid accumulation in muscle influences insulin sensitivity. Here we show that muscle-specific CGI-58 knockout mice relative to their controls have increased glucose tolerance and insulin sensitivity on a Western-type high-fat diet, despite TG accumulation in both heart and oxidative skeletal muscle and cholesterol deposition in heart. Although the intracardiomyocellular lipid deposition results in cardiac ventricular fibrosis and systolic dysfunction, muscle-specific CGI-58 knockout mice show increased glucose uptake in heart and soleus muscle, improved insulin signaling in insulin-sensitive tissues, and reduced plasma concentrations of glucose, insulin, and cholesterol. Hepatic contents of TG and cholesterol are also decreased in these animals. Cardiac steatosis is attributable, at least in part, to decreases in cardiac TG hydrolase activity and peroxisome proliferator-activated receptor-α/peroxisome proliferator-activated receptor-γ coactivator-1-dependent mitochondrial fatty acid oxidation. In conclusion, muscle CGI-58 deficiency causes cardiac dysfunction and fat deposition in oxidative muscles but induces a series of favorable metabolic changes in mice fed a high-fat diet.
Collapse
Affiliation(s)
- Ping Xie
- Departments of Biochemistry (P.X., Y.M., F.G., L.Y.) and Anesthesiology (L.G.), Wake Forest School of Medicine, Winston-Salem, North Carolina 27157; Department of Animal and Avian Sciences (A.K.G.G., Y.M., L.Y.), University of Maryland, College Park, Maryland 20742; Diabetes and Obesity Research Center (X.H., M.W.), Sanford-Burnham Medical Research Institute, Orlando, Florida 32827; Department of Biology (B.X., H.S.), Georgia State University, Atlanta, Georgia 30303; and The Key Laboratory of Remodeling-Related Cardiovascular Diseases (H.L.), Capital Medical University, Ministry of Education, Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing Anzhen Hospital Affiliated the Capital Medical University, Beijing 100029, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Khan SA, Sathyanarayan A, Mashek MT, Ong KT, Wollaston-Hayden EE, Mashek DG. ATGL-catalyzed lipolysis regulates SIRT1 to control PGC-1α/PPAR-α signaling. Diabetes 2015; 64:418-26. [PMID: 25614670 PMCID: PMC4303962 DOI: 10.2337/db14-0325] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Sirtuin 1 (SIRT1), an NAD(+)-dependent protein deacetylase, regulates a host of target proteins, including peroxisome proliferator-activated receptor (PPAR)-γ coactivator-1α (PGC-1α), a transcriptional coregulator that binds to numerous transcription factors in response to deacetylation to promote mitochondrial biogenesis and oxidative metabolism. Our laboratory and others have shown that adipose triglyceride lipase (ATGL) increases the activity of the nuclear receptor PPAR-α, a PGC-1α binding partner, to promote fatty acid oxidation. Fatty acids bind and activate PPAR-α; therefore, it has been presumed that fatty acids derived from ATGL-catalyzed lipolysis act as PPAR-α ligands. We provide an alternate mechanism that links ATGL to PPAR-α signaling. We show that SIRT1 deacetylase activity is positively regulated by ATGL to promote PGC-1α signaling. In addition, ATGL mediates the effects of β-adrenergic signaling on SIRT1 activity, and PGC-1α and PPAR-α target gene expression independent of changes in NAD(+). Moreover, SIRT1 is required for the induction of PGC-1α/PPAR-α target genes and oxidative metabolism in response to increased ATGL-mediated lipolysis. Taken together, this work identifies SIRT1 as a critical node that links β-adrenergic signaling and lipolysis to changes in the transcriptional regulation of oxidative metabolism.
Collapse
Affiliation(s)
- Salmaan Ahmed Khan
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN
| | | | - Mara T Mashek
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN
| | - Kuok Teong Ong
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN
| | | | - Douglas G Mashek
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN
| |
Collapse
|