1
|
Lu X, Ma H, Liu Y, Chen M, Dang J, Su X, Zhao Y, Wang K, Yang G, Zhang G, Li X, Gao A, Wang Y. Rhodotorula Yeast Culture Improved the Antioxidant Capacity, Lipid Metabolism, and Immunity of Sheep Livers. Vet Sci 2025; 12:314. [PMID: 40284815 PMCID: PMC12030957 DOI: 10.3390/vetsci12040314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/18/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
There is currently no research on the application evaluation of R. mucilaginosa yeast culture (RYC) in animal production. Therefore, this study investigated the effects of RYC on the antioxidant capacity, lipid metabolism, and immunity of sheep livers. Twenty-four 3-month-old Duhan male sheep (36 ± 4 kg) were divided into four groups. The control group received a basal diet, the L group received a basal diet + 10 g/sheep/day RYC, the M group received a basal diet + 20 g/sheep/day RYC, and the H group received a basal diet + 40 g/sheep/day RYC. The trial lasted for 75 days. The results showed that the content of glutathione peroxidase in the livers of sheep in group M was significantly increased by 26.6%, and the content of malondialdehyde was significantly decreased by 38% (p < 0.05). Additionally, the serum levels of total cholesterol, triglycerides, high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol significantly decreased; the liver content of C16:0 decreased; and the levels of C18:2n6C and C20:1 increased (p < 0.05). Furthermore, the contents of cytokines TNF-α and IFN-γ in sheep livers from the M group were also significantly decreased by 20% and 24.8%, respectively (p < 0.05). These findings suggest that supplementation with 20 g/sheep/day RYC can enhance antioxidant capacity, improve lipid metabolism, and reduce inflammation in sheep livers, which is advantageous for farming healthy sheep.
Collapse
Affiliation(s)
- Xinyu Lu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Huiru Ma
- Hetao College, Bayannur 015000, China
| | - Yeqing Liu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Meiru Chen
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Jianlong Dang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Xiangtan Su
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Yahui Zhao
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Ke Wang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Guang Yang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Gaowei Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Xiaorui Li
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Aiqin Gao
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Yuan Wang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Herbivorous Livestock Feed Engineering Technology Research Center, Hohhot 010018, China
| |
Collapse
|
2
|
Steinhoff JS, Wagner C, Dähnhardt HE, Košić K, Meng Y, Taschler U, Pajed L, Yang N, Wulff S, Kiefer MF, Petricek KM, Flores RE, Li C, Dittrich S, Sommerfeld M, Guillou H, Henze A, Raila J, Wowro SJ, Schoiswohl G, Lass A, Schupp M. Adipocyte HSL is required for maintaining circulating vitamin A and RBP4 levels during fasting. EMBO Rep 2024; 25:2878-2895. [PMID: 38769419 PMCID: PMC11239848 DOI: 10.1038/s44319-024-00158-x] [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: 03/27/2024] [Revised: 04/19/2024] [Accepted: 04/30/2024] [Indexed: 05/22/2024] Open
Abstract
Vitamin A (retinol) is distributed via the blood bound to its specific carrier protein, retinol-binding protein 4 (RBP4). Retinol-loaded RBP4 is secreted into the circulation exclusively from hepatocytes, thereby mobilizing hepatic retinoid stores that represent the major vitamin A reserves in the body. The relevance of extrahepatic retinoid stores for circulating retinol and RBP4 levels that are usually kept within narrow physiological limits is unknown. Here, we show that fasting affects retinoid mobilization in a tissue-specific manner, and that hormone-sensitive lipase (HSL) in adipose tissue is required to maintain serum concentrations of retinol and RBP4 during fasting in mice. We found that extracellular retinol-free apo-RBP4 induces retinol release by adipocytes in an HSL-dependent manner. Consistently, global or adipocyte-specific HSL deficiency leads to an accumulation of retinoids in adipose tissue and a drop of serum retinol and RBP4 during fasting, which affects retinoid-responsive gene expression in eye and kidney and lowers renal retinoid content. These findings establish a novel crosstalk between liver and adipose tissue retinoid stores for the maintenance of systemic vitamin A homeostasis during fasting.
Collapse
Affiliation(s)
- Julia S Steinhoff
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Carina Wagner
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Henriette E Dähnhardt
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Kristina Košić
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Yueming Meng
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Ulrike Taschler
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Laura Pajed
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Na Yang
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Sascha Wulff
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Marie F Kiefer
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Konstantin M Petricek
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Roberto E Flores
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Chen Li
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Sarah Dittrich
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Manuela Sommerfeld
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Hervé Guillou
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, Toulouse, France
| | - Andrea Henze
- Martin Luther University Halle-Wittenberg, Institute of Agricultural and Nutritional Sciences, Halle, Germany
- Junior Research Group ProAID, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Jens Raila
- Department of Physiology and Pathophysiology, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Sylvia J Wowro
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Gabriele Schoiswohl
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Achim Lass
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| | - Michael Schupp
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany.
| |
Collapse
|
3
|
Chen X, Lu T, Zheng Y, Lin Z, Liu C, Yuan D, Yuan C. miR-155-5p promotes hepatic steatosis via PICALM-mediated autophagy in aging hepatocytes. Arch Gerontol Geriatr 2024; 120:105327. [PMID: 38237377 DOI: 10.1016/j.archger.2024.105327] [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: 11/19/2023] [Revised: 12/31/2023] [Accepted: 01/08/2024] [Indexed: 03/10/2024]
Abstract
BACKGROUND Hepatic steatosis, a lipid disorder characterized by the accumulation of intrahepatic fat, is more prevalent in the elderly population. This study investigates the role of miR-155-5p in the autophagy dysregulation of aging hepatic steatosis. METHODS We established an aging mouse model in vivo and a hepatocellular senescence model induced by low serum and palmitic acid in vitro. The fluctuations of microRNAs were derived from RNA-seq data and confirmed by qPCR in 4- and 18-month-old mouse liver tissues. Hematoxylin-eosin (H&E) staining observed pathological changes. Markers of senescence, autophagy, and lipolysis genes were analyzed using Western blot and qPCR. Bioinformatics analysis predicted miR-155-5p's target gene PICALM, confirmed by dual luciferase reporter assay and transfection of miR-155-5p mimic/inhibitor into senescent hepatocytes. RESULTS Senescent markers (p21, p16, and p-P53) and miR-155-5p were up-regulated in aging liver tissues and senescent hepatocytes. Bioinformatics analysis identified PICALM as a target gene of miR-155-5p, a finding further supported by dual luciferase reporter assays. Inhibition of miR-155-5p reduced expression of senescent marker genes (p16, p21, p-P53), improved autophagy (evidenced by increased LC3B-II and ATG5, and decreased P62), and enhanced lipolysis (indicated by increased ATGL and p-HSL) in senescent hepatocytes. Oil red O staining confirmed that miR-155-5p inhibition significantly reduced lipid accumulation in these cells. CONCLUSIONS This study suggests a potential new therapeutic approach for age-related hepatic steatosis through the inhibition of miR-155-5p to enhance autophagy.
Collapse
Affiliation(s)
- Xiaoling Chen
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, HuBei, 443002, China
| | - Ting Lu
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, HuBei, 443002, China
| | - Ying Zheng
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, HuBei, 443002, China
| | - Zhiyong Lin
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, HuBei, 443002, China
| | - Chaoqi Liu
- Tumor Microenvironment and Immunotherapy Key Laboratory of Hubei province in China, China Three Gorges University, School of Medicine, Yichang, 443002, China; College of Basic Medical Science, China Three Gorges University, Yichang, HuBei, 443002, China.
| | - Ding Yuan
- College of Medicine and Health Science, China Three Gorges University, Yichang, HuBei, 443002, China.
| | - Chengfu Yuan
- College of Basic Medical Science, China Three Gorges University, Yichang, HuBei, 443002, China; Third Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, China Three Gorges University, School of Medicine, Yichang, 443002, China.
| |
Collapse
|
4
|
Ye J, Gong M, Zhang Y, Xu Q, Zhao J. Effects of Fermented Extracts of Wuniuzao Dark Loose Tea on Hepatic Sterol Regulatory Element-Binding Protein Pathway and Gut Microbiota Disorder in Obese Mice. J Nutr 2024; 154:626-637. [PMID: 38110182 DOI: 10.1016/j.tjnut.2023.12.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 12/04/2023] [Accepted: 12/13/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Artificially fermented dark loose tea is a type of novel dark tea prepared via fermentation by Eurotium cristatum. The effects of artificially fermented dark loose tea on lipid metabolism are still unclear. OBJECTIVES This study aimed to explore if artificially fermented dark loose tea has the same effects as naturally fermented dark loose tea in regulating hepatic lipid metabolism. METHODS Thirty-six 8-wk-old male C57BL/6 mice were randomly divided into 6 treatment groups, including normal control (NC), high-fat diet (HFD), positive control (PC), Wuniuzao dark raw tea (WDT), Wuniuzao naturally fermented dark loose tea (NFLT), and Wuniuzao artificially fermented dark loose tea (AFLT) groups. The HFD, PC, WDT, NFLT, and AFLT groups were fed a HFD. The PC group was supplemented with atorvastatin (10 mg/kg). The WDT group was supplemented with WDT (300 mg/kg), the NFLT group with NFLT (300 mg/kg), and the AFLT group with AFLT (300 mg/kg). RESULTS The study compared the effect of WDT, NFLT, and AFLT on liver steatosis and gut microbiota disorder in obese mice. All 3 tea extracts reduced body weight, glucose tolerance, and serum lipid concentrations. Via sterol-regulatory element binding protein (SREBP)-mediated lipid metabolism, all 3 tea extracts alleviated hepatic steatosis in mice with obesity. Furthermore, NFLT and AFLT intervened in the abundance of Firmicutes, Bacteroidetes, Clostridia, Muribaculaceae, and Lachnospiraceae. CONCLUSION In mice with obesity induced by a HFD, WDT, NFLT, and AFLT may improve hepatic steatosis through an SREBP-mediated lipid metabolism. Moreover, NFLT and AFLT improved the composition of gut microbiota.
Collapse
Affiliation(s)
- Jiangcheng Ye
- Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, Institute of Food Nutrition and Quality Safety, College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Mingxiu Gong
- Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, Institute of Food Nutrition and Quality Safety, College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Yifan Zhang
- Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, Institute of Food Nutrition and Quality Safety, College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Qianqian Xu
- Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, Institute of Food Nutrition and Quality Safety, College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Jin Zhao
- Key Laboratory of Specialty Agri-product Quality and Hazard Controlling Technology of Zhejiang Province, Institute of Food Nutrition and Quality Safety, College of Life Sciences, China Jiliang University, Hangzhou, China.
| |
Collapse
|
5
|
Cho CH, Patel S, Rajbhandari P. Adipose tissue lipid metabolism: lipolysis. Curr Opin Genet Dev 2023; 83:102114. [PMID: 37738733 DOI: 10.1016/j.gde.2023.102114] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/15/2023] [Accepted: 08/23/2023] [Indexed: 09/24/2023]
Abstract
White adipose tissue stores fatty acid (FA) as triglyceride in the lipid droplet organelle of highly specialized cells known as fat cells or adipocytes. Depending on the nutritional state and energy demand, hormonal and biochemical signals converge on activating an elegant and fundamental process known as lipolysis, which involves triglyceride hydrolysis to FAs. Almost six decades of work have vastly expanded our knowledge of lipolysis from enzymatic processes to complex protein assembly, disassembly, and post-translational modification. Research in recent decades ushered in the discovery of new lipolytic enzymes and coregulators and the characterization of numerous factors and signaling pathways that regulate lipid hydrolysis on transcriptional and post-transcriptional levels. This review will discuss recent developments with particular emphasis on the past two years in enzymatic lipolytic pathways and transcriptional regulation of lipolysis. We will summarize the positive and negative regulators of lipolysis, the adipose tissue microenvironment in lipolysis, and the systemic effects of lipolysis. The dynamic nature of adipocyte lipolysis is emerging as an essential regulator of metabolism and energy balance, and we will discuss recent developments in this area.
Collapse
Affiliation(s)
- Chung Hwan Cho
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sanil Patel
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Prashant Rajbhandari
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Diabetes, Obesity, and Metabolism Institute, Department of Endocrinology and Bone Disease, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place New York, NY 10029 USA.
| |
Collapse
|
6
|
Hu T, Li Z, Gong C, Xiong Y, Sun S, Xing J, Li Y, Li R, Wang Y, Wang Y, Lin Y. FOS Inhibits the Differentiation of Intramuscular Adipocytes in Goats. Genes (Basel) 2023; 14:2088. [PMID: 38003034 PMCID: PMC10671551 DOI: 10.3390/genes14112088] [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: 09/27/2023] [Revised: 11/14/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Goat intramuscular fat (IMF) deposition is precisely regulated by many key genes as well as transcription factors. Nevertheless, the potential of the regulators of goat IMF deposition remains undefined. In this work, we reported that the transcription factor FOS is expressed at a low level at the early differentiation stage and at a high level in late differentiation. The overexpression of FOS inhibited intramuscular adipocyte lipid accumulation and significantly downregulated the expressions of PPARγ, C/EBPβ, C/EBPα, AP2, SREBP1, FASN, ACC, HSL, and ATGL. Consistently, the knockdown of FOS, facilitated by two distinct siRNAs, significantly promoted intramuscular adipocyte lipid accumulation. Moreover, our analysis revealed multiple potential binding sites for FOS on the promoters of PPARγ, C/EBPβ, and C/EBPα. The expression changes in PPARγ, C/EBPβ, and C/EBPα during intramuscular adipogenesis were opposite to that of FOS. In summary, FOS inhibits intramuscular lipogenesis in goats and potentially negatively regulates the expressions of PPARγ, C/EBPβ, and C/EBPα genes. Our research will provide valuable data for the underlying molecular mechanism of the FOS regulation network of intramuscular lipogenesis.
Collapse
Affiliation(s)
- Tingting Hu
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| | - Zhibin Li
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| | - Chengsi Gong
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| | - Yan Xiong
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| | - Shiyu Sun
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
| | - Jiani Xing
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| | - Yanyan Li
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| | - Ruiwen Li
- Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 611731, China;
| | - Youli Wang
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| | - Yong Wang
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| | - Yaqiu Lin
- College of Animal Science and Veterinary, Southwest Minzu University, Chengdu 610041, China; (T.H.); (Z.L.); (C.G.); (Y.X.); (S.S.); (J.X.); (Y.L.); (Y.W.); (Y.W.)
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| |
Collapse
|
7
|
Kolleritsch S, Pajed L, Tilp A, Hois V, Pototschnig I, Kien B, Diwoky C, Hoefler G, Schoiswohl G, Haemmerle G. Adverse cardiac remodeling augments adipose tissue ß-adrenergic signaling and lipolysis counteracting diet-induced obesity. J Biol Chem 2023; 299:104788. [PMID: 37150323 PMCID: PMC10318461 DOI: 10.1016/j.jbc.2023.104788] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 03/06/2023] [Accepted: 04/25/2023] [Indexed: 05/09/2023] Open
Abstract
Cardiac triacylglycerol accumulation is a common characteristic of obesity and type 2 diabetes and strongly correlates with heart morbidity and mortality. We have previously shown that cardiomyocyte-specific perilipin 5 overexpression (Plin5-Tg) provokes significant cardiac steatosis via lowering cardiac lipolysis and fatty acid (FA) oxidation. In strong contrast to cardiac steatosis and lethal heart dysfunction in adipose triglyceride lipase deficiency, Plin5-Tg mice do not develop heart dysfunction and show a normal life span on chow diet. This finding prompted us to study heart function and energy metabolism in Plin5-Tg mice fed high-fat diet (HFD). Plin5-Tg mice showed adverse cardiac remodeling on HFD with heart function only being compromised in one-year-old mice, likely due to reduced cardiac FA uptake, thereby delaying deleterious cardiac lipotoxicity. Notably, Plin5-Tg mice were less obese and protected from glucose intolerance on HFD. Changes in cardiac energy catabolism in Plin5-Tg mice increased ß-adrenergic signaling, lipolytic, and thermogenic protein expression in adipose tissue ultimately counteracting HFD-induced obesity. Acute cold exposure further augmented ß-adrenergic signaling in Plin5-Tg mice, whereas housing at thermoneutrality did not protect Plin5-Tg mice from HFD-induced obesity albeit blood glucose and insulin levels remained low in transgenic mice. Overall, our data suggest that the limited capacity for myocardial FA oxidation on HFD increases cardiac stress in Plin5-Tg mice, thereby stimulating adipose tissue ß-adrenergic signaling, triacylglycerol catabolism, and thermogenesis. However, long-term HFD-mediated metabolic stress causes contractile dysfunction in Plin5-Tg mice, which emphasizes the importance of a carefully controlled dietary regime in patients with cardiac steatosis and hypertrophy.
Collapse
Affiliation(s)
| | - Laura Pajed
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Anna Tilp
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Victoria Hois
- Division of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | | | - Benedikt Kien
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Clemens Diwoky
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Gerald Hoefler
- Diagnostic & Research Institute of Pathology, Medical University of Graz, Graz, Austria; BioTechMed, Graz, Graz, Austria
| | - Gabriele Schoiswohl
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; BioTechMed, Graz, Graz, Austria; Department of Pharmacology and Toxicology, University of Graz, Graz, Austria.
| | - Guenter Haemmerle
- Institute of Molecular Biosciences, University of Graz, Graz, Austria; BioTechMed, Graz, Graz, Austria.
| |
Collapse
|
8
|
Rosiglitazone Reverses Inflammation in Epididymal White Adipose Tissue in Hormone-Sensitive Lipase-Knockout Mice. J Lipid Res 2022; 64:100305. [PMID: 36273647 PMCID: PMC9760656 DOI: 10.1016/j.jlr.2022.100305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/05/2022] [Accepted: 09/25/2022] [Indexed: 11/06/2022] Open
Abstract
Hormone-sensitive lipase (HSL) plays a crucial role in intracellular lipolysis, and loss of HSL leads to diacylglycerol (DAG) accumulation, reduced FA mobilization, and impaired PPARγ signaling. Hsl knockout mice exhibit adipose tissue inflammation, but the underlying mechanisms are still not clear. Here, we investigated if and to what extent HSL loss contributes to endoplasmic reticulum (ER) stress and adipose tissue inflammation in Hsl knockout mice. Furthermore, we were interested in how impaired PPARγ signaling affects the development of inflammation in epididymal white adipose tissue (eWAT) and inguinal white adipose tissue (iWAT) of Hsl knockout mice and if DAG and ceramide accumulation contribute to adipose tissue inflammation and ER stress. Ultrastructural analysis showed a markedly dilated ER in both eWAT and iWAT upon loss of HSL. In addition, Hsl knockout mice exhibited macrophage infiltration and increased F4/80 mRNA expression, a marker of macrophage activation, in eWAT, but not in iWAT. We show that treatment with rosiglitazone, a PPARγ agonist, attenuated macrophage infiltration and ameliorated inflammation of eWAT, but expression of ER stress markers remained unchanged, as did DAG and ceramide levels in eWAT. Taken together, we show that HSL loss promoted ER stress in both eWAT and iWAT of Hsl knockout mice, but inflammation and macrophage infiltration occurred mainly in eWAT. Also, PPARγ activation reversed inflammation but not ER stress and DAG accumulation. These data indicate that neither reduction of DAG levels nor ER stress contribute to the reversal of eWAT inflammation in Hsl knockout mice.
Collapse
|
9
|
Role of Distinct Fat Depots in Metabolic Regulation and Pathological Implications. Rev Physiol Biochem Pharmacol 2022; 186:135-176. [PMID: 35915363 DOI: 10.1007/112_2022_73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
People suffering from obesity and associated metabolic disorders including diabetes are increasing exponentially around the world. Adipose tissue (AT) distribution and alteration in their biochemical properties play a major role in the pathogenesis of these diseases. Emerging evidence suggests that AT heterogeneity and depot-specific physiological changes are vital in the development of insulin resistance in peripheral tissues like muscle and liver. Classically, AT depots are classified into white adipose tissue (WAT) and brown adipose tissue (BAT); WAT is the site of fatty acid storage, while BAT is a dedicated organ of metabolic heat production. The discovery of beige adipocyte clusters in WAT depots indicates AT heterogeneity has a more central role than hither to ascribed. Therefore, we have discussed in detail the current state of understanding on cellular and molecular origin of different AT depots and their relevance toward physiological metabolic homeostasis. A major focus is to highlight the correlation between altered WAT distribution in the body and metabolic pathogenesis in animal models and humans. We have also underscored the disparity in the molecular (including signaling) changes in various WAT tissues during diabetic pathogenesis. Exercise-mediated beneficial alteration in WAT physiology/distribution that protects against metabolic disorders is evolving. Here we have discussed the depot-specific biochemical adjustments induced by different forms of exercise. A detailed understanding of the molecular details of inter-organ crosstalk via substrate utilization/storage and signaling through chemokines provide strategies to target selected WAT depots to pharmacologically mimic the benefits of exercise countering metabolic diseases including diabetes.
Collapse
|
10
|
Grabner GF, Xie H, Schweiger M, Zechner R. Lipolysis: cellular mechanisms for lipid mobilization from fat stores. Nat Metab 2021; 3:1445-1465. [PMID: 34799702 DOI: 10.1038/s42255-021-00493-6] [Citation(s) in RCA: 363] [Impact Index Per Article: 90.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/15/2021] [Indexed: 12/13/2022]
Abstract
The perception that intracellular lipolysis is a straightforward process that releases fatty acids from fat stores in adipose tissue to generate energy has experienced major revisions over the last two decades. The discovery of new lipolytic enzymes and coregulators, the demonstration that lipophagy and lysosomal lipolysis contribute to the degradation of cellular lipid stores and the characterization of numerous factors and signalling pathways that regulate lipid hydrolysis on transcriptional and post-transcriptional levels have revolutionized our understanding of lipolysis. In this review, we focus on the mechanisms that facilitate intracellular fatty-acid mobilization, drawing on canonical and noncanonical enzymatic pathways. We summarize how intracellular lipolysis affects lipid-mediated signalling, metabolic regulation and energy homeostasis in multiple organs. Finally, we examine how these processes affect pathogenesis and how lipolysis may be targeted to potentially prevent or treat various diseases.
Collapse
Affiliation(s)
- Gernot F Grabner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Hao Xie
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Martina Schweiger
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|