1
|
Ge Y, Zhan H, Wu S, Wang J, Xu Y, Liang Y, Peng L, Gao L, Zhao J, He Z. GPR40 signaling in agouti-related peptide neurons mediates fat preference. Life Sci 2025; 373:123677. [PMID: 40320138 DOI: 10.1016/j.lfs.2025.123677] [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: 02/25/2025] [Revised: 04/18/2025] [Accepted: 05/01/2025] [Indexed: 05/17/2025]
Abstract
AIMS Fat preference is mediated by fatty acid receptors in the oral, intestinal, and central nervous systems, but their central nervous system roles remain unclear. Here, we investigated how GPR40, a medium- and long-chain fatty acid receptor, regulates fat preference via agouti-related peptide (AgRP) neurons in the hypothalamic arcuate nucleus (ARC). MATERIALS AND METHODS AgRP neuron-specific Gpr40 knockout mice were generated to investigate the role of GPR40 in dietary fat preference. Behavioral tests were conducted to assess dietary preferences, and metabolic analyses were performed after starvation. We also measured the activity of AgRP neurons and the expression levels of AgRP and neuropeptide Y (NPY) to explore the mechanisms. KEY FINDINGS Our results indicate that GPR40 is a novel signaling pathway that regulates fat preference in hypothalamic AgRP neurons, but not in pro-opiomelanocortin (POMC) neurons. AgRP-specific Gpr40 knockout mice displayed a reduced preference for fat. This alteration in dietary preference was not associated with behavioral anomalies such as anxiety, depression, or deficits in short-term memory. Additionally, Gpr40 deletion in ARC AgRP neurons resulted in a diminished metabolic state, increased AgRP neuronal activity, and elevated levels of AgRP and NPY peptides following starvation, leading to reduced fat intake and increased carbohydrate intake. Inhibition of AgRP neuronal activity in AgRP-specific Gpr40 knockout mice rescued the observed changes in fat preference. SIGNIFICANCE GPR40 signaling in AgRP neurons plays a critical role in regulating fat preference by modulating neuronal activity and the expression of AgRP and NPY peptides.
Collapse
Affiliation(s)
- Yueping Ge
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration and Practice Center, Shandong University, Jinan, Shandong 250021, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism; Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Huidong Zhan
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration and Practice Center, Shandong University, Jinan, Shandong 250021, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism; Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Shanshan Wu
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration and Practice Center, Shandong University, Jinan, Shandong 250021, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism; Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Jing Wang
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration and Practice Center, Shandong University, Jinan, Shandong 250021, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism; Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Yang Xu
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration and Practice Center, Shandong University, Jinan, Shandong 250021, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism; Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Yixiao Liang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism; Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Li Peng
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration and Practice Center, Shandong University, Jinan, Shandong 250021, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism; Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Ling Gao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism; Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Jiajun Zhao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism; Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Zhao He
- Department of Endocrinology, Shandong Provincial Hospital & Medical Integration and Practice Center, Shandong University, Jinan, Shandong 250021, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China; Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Shandong Key Laboratory of Endocrinology and Lipid Metabolism; Shandong Institute of Endocrine and Metabolic Diseases, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| |
Collapse
|
2
|
Cao B, Zhou J, Xia B, Li X, Wang R, Xu Y, Li C. Therapeutic potential of a choline-zinc-vitamin E nutraceutical complex in ameliorating thioacetamide-induced nonalcoholic fatty liver pathology in zebrafish. PLoS One 2025; 20:e0324164. [PMID: 40392901 PMCID: PMC12091810 DOI: 10.1371/journal.pone.0324164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 04/21/2025] [Indexed: 05/22/2025] Open
Abstract
Choline has been proven to be effective in maintaining liver function. However, the effect of choline, in combination with other nutrients, on the improvement of non-alcoholic fatty liver disease (NAFLD) remains unclear. This study aimed to investigate the potential effect of the nutraceutical complex containing choline bitartrate, zinc citrate, and dl-α-Tocopheryl acetate on NAFLD in the zebrafish model. The NAFLD model was induced in zebrafish by administering thioacetamide. Experimental groups were established, including a normal control group, the model control group, the positive control group, the nutraceutical complex intervention group, and the choline bitartrate alone intervention group. The intervention was administered to the zebrafish in a water-soluble form, while the positive control group received polyene phosphatidylcholine at a concentration of 50.0 μg/mL. Notably, the protective effect of the nutraceutical complex against NAFLD is more pronounced than that observed with choline bitartrate supplementation alone. The results of transcriptomics and quantitative real-time PCR showed that the potential mechanisms underlying the effects of the nutraceutical complex might involve the upregulation of acacia, acsl1a, fbp2 gene expression, and the downregulation of tbc1d1 gene expression. These results were further validated by western blotting and overexpression experiments. Our findings indicated that choline bitartrate, zinc citrate, and dl-α-Tocopheryl acetate can help improve NAFLD. The results of this study provide evidence for the application of the nutraceutical complex in the improvement of NAFLD.
Collapse
Affiliation(s)
- Bingbing Cao
- Hunter Biotechnology, Inc., Hangzhou, Zhejiang, The People’s Republic of China
| | - Jiali Zhou
- Hunter Biotechnology, Inc., Hangzhou, Zhejiang, The People’s Republic of China
| | - Bo Xia
- Hunter Biotechnology, Inc., Hangzhou, Zhejiang, The People’s Republic of China
| | - Xiaoqing Li
- Opella, Shanghai, The People’s Republic of China
| | - Rui Wang
- Opella, Shanghai, The People’s Republic of China
| | - Yiqiao Xu
- Hunter Biotechnology, Inc., Hangzhou, Zhejiang, The People’s Republic of China
| | - Chunqi Li
- Hunter Biotechnology, Inc., Hangzhou, Zhejiang, The People’s Republic of China
| |
Collapse
|
3
|
He L, She X, Guo L, Gao M, Wang S, Lu Z, Guo H, Li R, Nie Y, Xing J, Ji L. Hepatic AKAP1 deficiency exacerbates diet-induced MASLD by enhancing GPAT1-mediated lysophosphatidic acid synthesis. Nat Commun 2025; 16:4286. [PMID: 40341440 PMCID: PMC12062205 DOI: 10.1038/s41467-025-58790-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 04/02/2025] [Indexed: 05/10/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), closely associated with obesity, can progress to metabolic dysfunction-associated steatohepatitis when the liver undergoes overt inflammatory damage. A-kinase anchoring protein 1 (AKAP1) has been shown to control lipid accumulation in brown adipocytes. However, the role of AKAP1 signaling in hepatic lipid metabolism and MASLD remains poorly understood. Here, we showed that hepatocyte-specific AKAP1 deficiency exacerbated hepatic steatosis and steatohepatitis in male mice subjected to a high-fat diet and fast-food diet, respectively. Mechanistically, AKAP1 directly phosphorylated and inactivated glycerol-3-phosphate acyltransferase 1 (GPAT1) in a PKA-dependent manner, thus suppressing lysophosphatidic acid (LPA) production. Increased endogenous LPA in hepatocytes promoted hepatocellular triglyceride (TG) synthesis and initiated pronounced inflammatory response in Kupffer cells. Restoring hepatic AKAP1 or repressing LPA levels via GPAT1 knockdown alleviated MASLD exacerbation. Overall, AKAP1 plays a protective role against MASLD by inhibiting GPAT1 activity, highlighting the potential of targeting AKAP1/PKA/GPAT1 signalosome for MASLD therapy.
Collapse
Affiliation(s)
- Linjie He
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiaojuan She
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
- College of Life Sciences, Yan'an University, Yan'an, Shaanxi, China
| | - Lifei Guo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
- College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Mingshu Gao
- College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
- National Demonstration Center for Experimental Basic Medical Science Education, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shuangbin Wang
- National Demonstration Center for Experimental Basic Medical Science Education, Fourth Military Medical University, Xi'an, Shaanxi, China
- Medical College of Yan'an University, Yan'an, Shaanxi, China
| | - Zhenxing Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Haitao Guo
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Renlong Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yongzhan Nie
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jinliang Xing
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Lele Ji
- National Demonstration Center for Experimental Basic Medical Science Education, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
4
|
Mokhtari S, Zarrin M, Samadian F, Ahmadpour A. Both Feed Restriction and Transition Period Suppressed Adipose Tissue MRNA Abundance of Genes Involved in Lipogenesis and Lipolysis in Fat-Tailed Ewes. J Anim Physiol Anim Nutr (Berl) 2025; 109:646-654. [PMID: 39696981 DOI: 10.1111/jpn.14085] [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: 01/31/2024] [Revised: 08/28/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024]
Abstract
This study sought to investigate the consequences of the induced feed restriction during the transition period on the mRNA abundance of genes entangled in lipogenesis and lipolysis in the tail adipose of fat-tailed sheep. Twenty fat-tailed ewes were randomised into the control (Control; n = 10) and restriction (Restriction; n = 10) groups. Control animals were fed 100% of the balanced diet pre-(Week -5 to parturition) and post-partum (parturition to Week 5). Restriction ewes received equivalent 100%, 50%, 65%, 80%, and 100% of the balanced ration at Weeks -5, -4, -3, -2, and -1 relative to parturition, respectively. After parturition, the latter group received the equivalent of 100%, 50%, 65%, 80%, and 100% of the diet recommendation at weeks 1, 2, 3, 4, and 5, respectively. At the end of weeks -3 and 3, tail adipose were sampled under local anesthesia. Fatty acid synthase, acetyl-CoA carboxylase, carnitine palmitoyltransferase I and II, and acyl-CoA synthase long-chain family member-1 mRNA abundances were measured using the TaqMan quantity real-time PCR. A mixed model procedure of SAS software was used to evaluate the results. Feed restriction downregulated target genes' mRNA abundance during both pre- and post-partum. Parturition suppressed the mRNA abundance of measured genes in both groups. Established on the outcomes, lipogenesis, and lipolysis of the adipose tissue would be influenced by metabolite and hormone instability during the transition period and feed restriction. The lessening of adipose tissue lipogenesis and lipolysis might be a hemostatic response to cope with the energy insufficiency for fetal growth and the onset of lactation and also prevent the induction of inflammation, metabolic disorders, and infectious diseases during feed restriction or transition period.
Collapse
Affiliation(s)
- Sara Mokhtari
- Department of Animal Science, Faculty of Agriculture, Yasouj University, Yasouj, Iran
| | - Mousa Zarrin
- Department of Animal Science, Faculty of Agriculture, Yasouj University, Yasouj, Iran
| | - Farhad Samadian
- Department of Animal Science, Faculty of Agriculture, Yasouj University, Yasouj, Iran
| | - Amir Ahmadpour
- Department of Animal Science, Faculty of Agriculture, Yasouj University, Yasouj, Iran
| |
Collapse
|
5
|
Gunasegaran B, Krishnamurthy S, Chow SS, Villanueva MD, Guller A, Ahn SB, Heng B. Comparative Analysis of HMC3 and C20 Microglial Cell Lines Reveals Differential Myeloid Characteristics and Responses to Immune Stimuli. Immunology 2025; 175:84-102. [PMID: 39961658 PMCID: PMC11982601 DOI: 10.1111/imm.13900] [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: 09/18/2024] [Revised: 12/11/2024] [Accepted: 01/07/2025] [Indexed: 04/11/2025] Open
Abstract
Microglia are the primary resident immune cells of the central nervous system (CNS) that respond to injury and infections. Being critical to CNS homeostasis, microglia also have been shown to contribute to neurodegenerative diseases and brain cancer. Hence, microglia are regarded as a potential therapeutic target in CNS diseases, resulting in an increased demand for reliable in vitro models. Two human microglia cell lines (HMC3 and C20) are being used in multiple in vitro studies, however, the knowledge of their biological and immunological characteristics remains limited. Our aim was to identify and compare the biological changes in these immortalised immune cells under normal physiological and immunologically challenged conditions. Using high-resolution quantitative mass spectrometry, we have examined in-depth proteomic profiles of non-stimulated and LPS or IFN-γ challenged HMC3 and C20 cells. Our findings reveal that HMC3 cells responded to both treatments through upregulation of immune, metabolic, and antiviral pathways, while C20 cells showed a response associated with mitochondrial and immune activities. Additionally, the secretome analysis demonstrated that both cell lines release IL-6 in response to LPS, while IFN-γ treatment resulted in altered kynurenine pathway activity, highlighting distinct immune and metabolic adaptations.
Collapse
Affiliation(s)
- Bavani Gunasegaran
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Shivani Krishnamurthy
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Sharron S. Chow
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Millijoy D. Villanueva
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
- Computational Neurosurgery (CNS) Lab, Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Anna Guller
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
- Computational Neurosurgery (CNS) Lab, Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Seong Beom Ahn
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Benjamin Heng
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| |
Collapse
|
6
|
Zhang P, Zhang H, Shahzad M, Kolachi HA, Li Y, Sheng H, Zhang X, Wan P, Zhao X. Supplementation of Forskolin and Linoleic Acid During IVC Improved the Developmental and Vitrification Efficiency of Bovine Embryos. Int J Mol Sci 2025; 26:4151. [PMID: 40362390 PMCID: PMC12071939 DOI: 10.3390/ijms26094151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
The success of assisted reproductive technology is contingent upon the growth potential of embryos post-vitrification process. When compared to in vivo embryos, it has been found that the high intracellular lipid accumulation inside the in vitro-derived embryos results in poor survival during vitrification. Based on this finding, the present study assessed the impact of incorporating forskolin and linoleic acid (FL) entering in vitro culture (IVC) on the embryos' cryo-survival, lipid content, and viability throughout vitrification. Lipid metabolomics and single-cell RNA sequencing (scRNA-seq) techniques were used to determine the underlying mechanism that the therapies were mimicking. It was observed that out of 726 identified lipids, 26 were expressed differentially between the control and FL groups, with 12 lipids upregulated and 14 lipids downregulated. These lipids were classified as Triacylglycerol (TG), Diacylglycerol (DG), Phosphatidylcholine (PC), and so on. A total of 1079 DEGs were detected between the FL and control groups, consisting of 644 upregulated genes and 435 downregulated genes. These DEGs were significantly enhanced in the arachidonic acid metabolism, lipolysis, fatty acid metabolism, cAMP signaling pathway, and other critical developmental pathways. Based on the observation, it was concluded that forskolin and linoleic acid decreased the droplet content of embryos by modulating lipid metabolism, thus enhancing the vitrified bovine embryos' cryo-survival.
Collapse
Affiliation(s)
- Peipei Zhang
- State Key Laboratory of Sheep Genetic Improvement and Healthy Breeding, Institute of Animal Husbandry and Veterinary Sciences, Xinjiang Academy of Agricultural and Reclamation Sciences, Shihezi 832061, China;
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Y.L.); (H.S.)
| | - Hang Zhang
- Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), No. 2 Yuanmingyuan Western Road, Haidian District, Beijing 100193, China; (H.Z.); (M.S.); (H.A.K.)
| | - Muhammad Shahzad
- Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), No. 2 Yuanmingyuan Western Road, Haidian District, Beijing 100193, China; (H.Z.); (M.S.); (H.A.K.)
| | - Hubdar Ali Kolachi
- Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), No. 2 Yuanmingyuan Western Road, Haidian District, Beijing 100193, China; (H.Z.); (M.S.); (H.A.K.)
| | - Yupeng Li
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Y.L.); (H.S.)
| | - Hui Sheng
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Y.L.); (H.S.)
| | - Xiaosheng Zhang
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Y.L.); (H.S.)
| | - Pengcheng Wan
- State Key Laboratory of Sheep Genetic Improvement and Healthy Breeding, Institute of Animal Husbandry and Veterinary Sciences, Xinjiang Academy of Agricultural and Reclamation Sciences, Shihezi 832061, China;
| | - Xueming Zhao
- Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), No. 2 Yuanmingyuan Western Road, Haidian District, Beijing 100193, China; (H.Z.); (M.S.); (H.A.K.)
| |
Collapse
|
7
|
Liu M, Zou DS, Zhang XY, Wang DH. Huddling behavior regulate adaptive thermogenesis in Brandt's voles (Lasiopodomys brandtii). Cell Biosci 2025; 15:51. [PMID: 40270019 PMCID: PMC12020165 DOI: 10.1186/s13578-025-01391-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 04/08/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Brown adipose tissue (BAT) is the main site of non-shivering thermogenesis (NST) in small mammals, playing an important role in maintaining body temperature and energy balance. Huddling is a behavioral strategy for small rodents to save energy and improve the survival under cold environments. However, the way of huddling behavior influence on hypothalamus, which regulate BAT thermogenesis in small mammals is rarely illustrated. We used male Brandt's voles (Lasiopodomys brandtii) to explore the possible regulation mechanisms in BAT thermogenesis by the way of cold acclimation and huddling behavior. RESULTS There is a strong relationship between huddling behavior and NST in BAT. The hypothalamus, which is impacted by huddling behavior, influences PPAR signaling pathway in the BAT, and induces thermogenesis through Calcium signaling pathway. PPAR pathway causes crosstalk among NF-κB signaling pathway, Thermogenesis and Fatty acid metabolism to perform functions for thermogenesis. CONCLUSIONS The results suggest that huddling behavior can modulate adaptive thermogenesis in BAT. Cold acclimation and huddling had a synergistic effect on the regulation of thermogenic function, the hypothalamus mediates thermogenic changes in BAT induced by huddling behavior. In BAT, the specific pathway of thermogenesis is as follows: TRAF6-PPARγ-UCP1-SUCLG1.
Collapse
Affiliation(s)
- Min Liu
- School of Life Sciences, Shandong University, Qingdao, 266237, China
| | - De-Sheng Zou
- School of Life Sciences, Shandong University, Qingdao, 266237, China
| | - Xue-Ying Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - De-Hua Wang
- School of Life Sciences, Shandong University, Qingdao, 266237, China.
| |
Collapse
|
8
|
Li J, Zou P, Xiao R, Wang Y. Indole-3-propionic acid alleviates DSS-induced colitis in mice through macrophage glycolipid metabolism. Int Immunopharmacol 2025; 152:114388. [PMID: 40086057 DOI: 10.1016/j.intimp.2025.114388] [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: 12/22/2024] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 03/16/2025]
Abstract
Ulcerative colitis (UC) is a chronic relapsing inflammatory bowel disease for which current therapeutic approaches still face many dilemmas, and targeting macrophage polarization and metabolism for the treatment of this disease is a potentially effective strategy. The gut microbial metabolite indole-3-propionic acid (IPA) has favorable anti-inflammatory and antioxidant effects and plays a role in a variety of disease models. IPA is effective in the treatment of UC, but the underlying mechanisms have not been well explored. In the present study, we investigated the mechanisms by which IPA ameliorates colitis in mice from the perspective of macrophage polarization and metabolism. In this study, mice colitis was induced by sodium dextran sulfate and treated with oral IPA. RAW264.7 cells were induced by LPS to polarize into M1 macrophages and treated with IPA. The results showed that IPA could improve colitis by inhibiting M1 polarization of colonic macrophages and promoting M2 polarization. The inhibition of IPA on M1 macrophages was verified in vitro through JNK/MAPK pathway, which inhibited the glycolysis of macrophages. IPA promotes macrophage M2 polarization and enhances fatty acid oxidation through upregulating of CPT1A and ACSL1, which may be related to the activation of PPAR-γ. In summary, IPA can improve colitis by regulating macrophage glucose and lipid metabolism, and targeting intestinal macrophage metabolism may be an effective target for the treatment of UC.
Collapse
Affiliation(s)
- Jiahong Li
- Beijing Children Hospital, Capital Medical University, Beijing 100045, China
| | - Peicen Zou
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Ruiqi Xiao
- Capital Institute of Pediatrics, Beijing 100020, China
| | - Yajuan Wang
- Children's Hospital, Capital Institute of Pediatrics, Beijing 100020, China.
| |
Collapse
|
9
|
Huang H, Liu L, Liang Z, Wang Q, Li C, Huang Z, Zhao Z, Han W. C-type natriuretic peptide regulates lipid metabolism through a NPRB-PPAR pathway in the intramuscular and subcutaneous adipocytes in chickens. Sci Rep 2025; 15:13018. [PMID: 40234429 PMCID: PMC12000514 DOI: 10.1038/s41598-025-86433-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: 08/12/2024] [Accepted: 01/10/2025] [Indexed: 04/17/2025] Open
Abstract
Natriuretic peptides (NPs) have an important role in lipid metabolism in skeletal muscle and adipose tissue in animals. C-type natriuretic peptide (CNP) is an important NP, but the molecular mechanisms that underlie its activity are not completely understood. Treatment of intramuscular fat (IMF) and subcutaneous fat (SCF) adipocytes with CNP led to decreased differentiation, promoted proliferation and lipolysis, and increased the expression of natriuretic peptide receptor B (NPRB) mRNA. Silencing natriuretic peptide C (NPPC) had the opposite results in IMF and SCF adipocytes. Transcriptome analysis found 665 differentially expressed genes (DEGs) in IMF adipocytes and 991 in SCF adipocytes. Seven genes in IMF adipocytes (FABP4, APOA1, ACOX2, ADIPOQ, CD36, FABP5, and LPL) and eight genes in SCF adipocytes (ACOX3, ACSL1, APOA1, CPT1A, CPT2, FABP4, PDPK1 and PPARα) are related to fat metabolism. Fifteen genes were found to be enriched in the peroxisome proliferator-activated receptor (PPAR) pathway. Integrated analysis identified 113 intersection genes in IMF and SCF adipocytes, two of which (APOA1 and FABP4) were enriched in the PPAR pathway. In conclusion, CNP may regulated lipid metabolism through the NPRB-PPAR pathway in both IMF and SCF adipocytes, FABP4 and APOA1 may be the key genes that mediated CNP regulation of fat deposition.
Collapse
Affiliation(s)
- Huayun Huang
- Institute of Poultry Science, Chinese Academy of Agriculture Sciences, 225125, Jiangsu, P. R. China
- College of Animal Science, Yangtze University, Jingzhou, 8060550, P. R. China
| | - Longzhou Liu
- Institute of Poultry Science, Chinese Academy of Agriculture Sciences, 225125, Jiangsu, P. R. China
- College of Animal Science, Yangtze University, Jingzhou, 8060550, P. R. China
| | - Zhong Liang
- Institute of Poultry Science, Chinese Academy of Agriculture Sciences, 225125, Jiangsu, P. R. China
- College of Animal Science, Yangtze University, Jingzhou, 8060550, P. R. China
| | - Qianbao Wang
- Institute of Poultry Science, Chinese Academy of Agriculture Sciences, 225125, Jiangsu, P. R. China
- College of Animal Science, Yangtze University, Jingzhou, 8060550, P. R. China
| | - Chunmiao Li
- Institute of Poultry Science, Chinese Academy of Agriculture Sciences, 225125, Jiangsu, P. R. China
- College of Animal Science, Yangtze University, Jingzhou, 8060550, P. R. China
| | - Zhengyang Huang
- Institute of Poultry Science, Chinese Academy of Agriculture Sciences, 225125, Jiangsu, P. R. China
- College of Animal Science, Yangtze University, Jingzhou, 8060550, P. R. China
| | - Zhenhua Zhao
- Institute of Poultry Science, Chinese Academy of Agriculture Sciences, 225125, Jiangsu, P. R. China.
- College of Animal Science, Yangtze University, Jingzhou, 8060550, P. R. China.
| | - Wei Han
- Institute of Poultry Science, Chinese Academy of Agriculture Sciences, 225125, Jiangsu, P. R. China.
- College of Animal Science, Yangtze University, Jingzhou, 8060550, P. R. China.
| |
Collapse
|
10
|
Preza S, Zheng B, Gao Z, Liu M, Biju A, Alvarez-Dominguez JR. DEC1 Regulates Human β Cell Functional Maturation and Circadian Rhythm. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.647023. [PMID: 40236051 PMCID: PMC11996484 DOI: 10.1101/2025.04.03.647023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Stem cell-derived islet (SC-islet) organoids offer hope for cell replacement therapy in diabetes, but their immature function remains a challenge. Mature islet function requires the β-cell circadian clock, yet how the clock regulates maturation is unclear. Here, we show that a circadian transcription factor specific to maturing SC-β cells, DEC1, regulates insulin responsiveness to glucose. SC-islet organoids form normally from DEC1 -ablated human pluripotent stem cells, but their insulin release capacity and glucose threshold fail to increase during in vitro culture and upon transplant. This deficit reflects downregulation of maturity-linked effectors of glucose utilization and insulin exocytosis, blunting glycolytic and oxidative metabolism, and is rescued by increasing metabolic flux. Moreover, DEC1 is needed to boost SC-islet maturity by synchronizing circadian glucose-responsive insulin secretion rhythms and clock machinery. Thus, DEC1 links circadian control to human β-cell maturation, highlighting its vitality to foster fully functional SC-islets.
Collapse
|
11
|
Wei M, Wang Y, Zhang Y, Qiao Y. Plin5: A potential therapeutic target for type 2 diabetes mellitus. Diabetol Metab Syndr 2025; 17:114. [PMID: 40176076 PMCID: PMC11963521 DOI: 10.1186/s13098-025-01680-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/22/2025] [Indexed: 04/04/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a kind of metabolic disease characterized by aberrant insulin secretion as a result of -cell loss or injury, or by impaired insulin sensitivity of peripheral tissues, which finally results in insulin resistance and a disturbance of glucose and lipid metabolism. Among them, lipid metabolism disorders lead to lipotoxicity through oxidative stress and inflammatory response, destroying the structure and function of tissues and cells. Abnormal lipid metabolism can lead to abnormal insulin signaling and disrupt glucose metabolism through a variety of pathways. Therefore, emphasizing lipid metabolism may be a crucial step in the prevention and treatment of T2DM. Plin5 is a lipid droplet surface protein, which can bi-directionally regulate lipid metabolism and plays an important role in lipolysis and fat synthesis. Plin5 can simultaneously decrease the buildup of free fatty acids in the cytoplasm, improve mitochondrial uptake of free fatty acids, speed up fatty acid oxidation through lipid drops-mitochondria interaction, and lessen lipotoxicity. In oxidative tissues like the heart, liver, and skeletal muscle, Plin5 is extensively expressed. And Plin5 is widely involved in β-cell apoptosis, insulin resistance, oxidative stress, inflammatory response and other pathological processes, which has important implications for exploring the pathogenesis of T2DM. In addition, recent studies have found that Plin5 is also closely related to T2DM and cancer, which provides a new perspective for exploring the relationship between T2DM and cancer.
Collapse
Affiliation(s)
- Mengjuan Wei
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yan Wang
- Beijing University of Chinese Medicine, Beijing, China
| | - Yufei Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yun Qiao
- Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
12
|
Venkatesh D, Sarkar S, Kandasamy T, Ghosh SS. In-silico identification and validation of Silibinin as a dual inhibitor for ENO1 and GLUT4 to curtail EMT signaling and TNBC progression. Comput Biol Chem 2025; 115:108312. [PMID: 39689434 DOI: 10.1016/j.compbiolchem.2024.108312] [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: 07/20/2024] [Revised: 11/29/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024]
Abstract
The aberrant metabolic reprogramming endows TNBC cells with sufficient ATP and lactate required for survival and metastasis. Hence, the intervention of the metabolic network represents a promising avenue to alleviate the Warburg effect in TNBC cells to impair their invasive and metastatic potential. Multitudinous in-silico analysis identified Enolase1 (ENO1) and the surface transporter protein, GLUT4 to be the potential targets for the abrogation of the metabolic network. The expression profiles of ENO1 and GLUT4 genes showed anomalous expression in various cancers, including breast cancer. Subsequently, the functional and physiological interactions of the target proteins were analyzed from the protein-protein interaction network. The pathway enrichment analysis identified the prime cancer signaling pathways in which these proteins are involved. Further, docking results bestowed Silibinin as the concurrent inhibitor of ENO1 and GLUT4. Moreover, the stable interaction of Silibinin with both proteins deciphered the binding free energies values of -48.86 and -104.31 KJ/mol from MMPBSA analysis and MD simulation, respectively. Furthermore, the cell viability, ROS assay, and live-dead imaging underscored the pronounced cytotoxicity of Silibinin, illuminating its capacity to incur apoptosis within TNBC cells. Additionally, glycolysis assay and gene expression analysis demonstrated the silibinin-mediated inhibition of the glycolysis pathway. Eventually, a lipidomic reprogramming towards fatty acid metabolism was established from the elevated lipid droplet accumulation, exogenous fatty acid uptake and de-novo lipogenesis. Nevertheless, repression of EMT and Wnt pathway progression by Silibinin was perceived from the gene expression studies. Overall, the current study highlights the tweaking of intricate signaling crosstalk between glycolysis and the Wnt pathway in TNBC cells through inhibiting ENO1 and GLUT4.
Collapse
Affiliation(s)
- Dheepika Venkatesh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India
| | - Shilpi Sarkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India
| | - Thirukumaran Kandasamy
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India
| | - Siddhartha Sankar Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India; Centre for Nanotechnology, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India.
| |
Collapse
|
13
|
Yuan X, Zhang X, Lin Y, Xie H, Wang Z, Hu X, Hu S, Li L, Liu H, He H, Han C, Gan X, Liao L, Xia L, Hu J, Wang J. Proteome of granulosa cells lipid droplets reveals mechanisms regulating lipid metabolism at hierarchical and pre-hierarchical follicle in goose. Front Vet Sci 2025; 12:1544718. [PMID: 40230795 PMCID: PMC11995638 DOI: 10.3389/fvets.2025.1544718] [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: 12/13/2024] [Accepted: 03/12/2025] [Indexed: 04/16/2025] Open
Abstract
Avian hierarchical follicles are formed by selection and dominance of pre-hierarchical follicles, and lipid metabolism plays a pivotal role in this process. The amount of lipid in goose follicular granulosa cells increases with the increase of culture time, and the neutral lipid in the cells is stored in the form of lipid droplets (LDs). LD-associated proteins (LDAPs) collaborate with LDs to regulate intracellular lipid homeostasis, which subsequently influences avian follicle development. The mechanism by which LDAPs regulate lipid metabolism in goose granulosa cells at different developmental stages is unclear. Therefore, using BODIPY staining, we found that at five time points during in vitro culture, the LD content in hierarchical granulosa cells was significantly higher than that in pre-hierarchical granulosa cells in this study (p < 0.001). Next, we identified LDAPs in both hierarchical and pre-hierarchical granulosa cells, and screened out 1,180, 922, 907, 663, and 1,313 differentially expressed proteins (DEPs) at the respective time points. Subsequently, by performing Clusters of Orthologous Groups (COGs) classification on the DEPs, we identified a large number of proteins related to lipid transport and metabolism. Following this, the potential functions of these DEPs were investigated through Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analysis. Finally, the important pathway of fatty acid degradation and the key protein ACSL3 were screened out using Short Time-series Expression Miner (STEM) and Protein-Protein Interaction (PPI) analysis methods. It is hypothesized that ACSL3 may potentially modulate lipid metabolism through the fatty acid degradation pathway, thereby contributing to the difference in lipid content between hierarchical and pre-hierarchical granulosa cells. These findings will provide a theoretical foundation for further studies on the role of LDs and LDAPs in avian follicle development.
Collapse
Affiliation(s)
- Xin Yuan
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Xi Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yueyue Lin
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Hengli Xie
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Zhujun Wang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Xinyue Hu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Shenqiang Hu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Liang Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Hehe Liu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Hua He
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Chunchun Han
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Xiang Gan
- Scientific Research Center, Guilin Medical University, Guilin, Guangxi, China
| | - Ling Liao
- College of Horticulture, Sichuan Agricultural University, Chengdu, China
| | - Lu Xia
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jiwei Hu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jiwen Wang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
14
|
Li B, Liu S, Han W, Song P, Sun H, Cao X, Di G, Chen P. Aquaporin five deficiency suppresses fatty acid oxidation and delays liver regeneration through the transcription factor PPAR. J Biol Chem 2025; 301:108303. [PMID: 39947476 PMCID: PMC11930093 DOI: 10.1016/j.jbc.2025.108303] [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: 10/07/2024] [Revised: 02/03/2025] [Accepted: 02/07/2025] [Indexed: 03/09/2025] Open
Abstract
After 70% partial hepatectomy (PHx), the metabolic pathways leading to hepatocyte lipid droplet accumulation during liver regeneration remain unclear. Aquaporin 5 (Aqp5) is an aquaporin that facilitates the transport of both water and hydrogen peroxide (H2O2). In this study, we observed delayed liver regeneration following PHx in Aqp5 knockout (Aqp5-/-) mice. Considering the role of Aqp5 in H2O2 transport, we hypothesized that deficiency in Aqp5 may induce oxidative stress and hepatocyte injury. Through the measurement of reactive oxygen species (ROS) and redox-related indices, we observed significant alterations in ROS levels as well as malondialdehyde (MDA), superoxide dismutase (SOD), and reduced glutathione (GSH) concentrations in regenerating livers lacking Aqp5 compared to wild-type controls. Oil Red O and 4-hydroxynonenal (4-HNE) staining results indicated that Aqp5 deficiency caused lipid accumulation during liver regeneration. The transcriptome sequencing results showed that the PPAR pathway is inhibited during the liver regeneration process in Aqp5 gene-knockout mice. The administration of the WY-14643 agonist, which targets the PPAR pathway, significantly mitigated delayed liver regeneration by enhancing hepatocyte proliferation and reducing lipid accumulation caused by Aqp5 deficiency. Our findings highlight the crucial role of Aqp5 in regulating H2O2 levels and lipid metabolism through the PPAR pathway during liver regeneration.
Collapse
Affiliation(s)
- Bin Li
- School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Shixu Liu
- School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Wenshuo Han
- School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Peirong Song
- School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Hetong Sun
- School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Xin Cao
- School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China.
| | - Guohu Di
- School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China; Institute of Stem Cell Regeneration Medicine, School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China.
| | - Peng Chen
- School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China; Institute of Stem Cell Regeneration Medicine, School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China; Department of Ophthalmology, Qingdao Eighth People's Hospital, Qingdao, Shandong Province, China.
| |
Collapse
|
15
|
Jiang S, Lin X, Chen B, Chen G, Kwan KJ, Liu J, Sun Q, Wang J, Lu Y, Tong J, Deng Y, Yu B, Tang J. ACSL1 Aggravates Thromboinflammation by LPC/LPA Metabolic Axis in Hyperlipidemia Associated Myocardial Ischemia-Reperfusion Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406359. [PMID: 39853712 PMCID: PMC11923997 DOI: 10.1002/advs.202406359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 12/24/2024] [Indexed: 01/26/2025]
Abstract
Acute myocardial infarction (AMI) is associated with well-established metabolic risk factors, especially hyperlipidemia and obesity. Myocardial ischemia-reperfusion injury (mIRI) significantly offsets the therapeutic efficacy of revascularization. Previous studies indicated that disrupted lipid homeostasis can lead to lipid peroxidation damage and inflammation, yet the underlying mechanisms remain unclear. Here, the study demonstrates that hyperlipidemia is a key driver of mIRI. Long-chain fatty acyl-CoA synthetase 1 (ACSL1) is upregulated in both hyperlipidemia and AMI patients. ACSL1 expression is induced by a high-fat microenvironment (oxLDL and palmitic acid) in a concentration-dependent manner. Interestingly, the protein level is positively correlated with total cholesterol level and thromboinflammatory biomarkers. Furthermore, ACSL1 reprogrammed lipid metabolism in monocytes, leading to the accumulation of lysophosphatidylcholine (LPC)/lysophosphatidic acid (LPA). The monocytic LPC/LPA axis accelerated lipid peroxidation and neutrophil extracellular traps (NETs)-induced thromboinflammation via the paracrine effect. The main LPA producer Autotaxinis is also induced under high-fat conditions and then exerts thromboinflammation response through converted LPC to LPA. Finally, ACSL1 knockdown or NETs release inhibitor (DNase I or GSK484) significantly alleviated mIRI in mice. These findings highlight ACSL1 and NETosis as potential key targets for preventing mIRI and underscore the lipid peroxidation in the mechanisms of ACSL1-mediated thromboinflammation.
Collapse
Affiliation(s)
- Shuai Jiang
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Xueguang Lin
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Bo Chen
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Gang Chen
- Department of CardiologyShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Kristine J.S. Kwan
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Jing Liu
- State Key Laboratory of Genetic EngineeringCollaborative Innovation Center for Genetics and DevelopmentSchool of Life Sciencesand Human Phenome InstituteFudan UniversityShanghai200438China
| | - Qi Sun
- Department of Endocrinology and MetabolismAffiliated Hospital of Nantong UniversityNantong226006China
| | - Jie Wang
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Yijie Lu
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Jindong Tong
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Ying Deng
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| | - Bo Yu
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
- Department of Vascular SurgeryHuashan HospitalFudan UniversityShanghai200040China
| | - Jingdong Tang
- Shanghai Key Laboratory of Vascular Lesions and RemodelingDepartment of Vascular SurgeryShanghai Pudong HospitalFudan University Pudong Medical CenterShanghai201399China
| |
Collapse
|
16
|
Astacio E, DiAngelo JR. The regulation of triglyceride storage by Acsx4 and Acsx5 in Drosophila fat tissue. MICROPUBLICATION BIOLOGY 2025; 2025:10.17912/micropub.biology.001430. [PMID: 39958912 PMCID: PMC11830124 DOI: 10.17912/micropub.biology.001430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/18/2025] [Accepted: 02/01/2025] [Indexed: 02/18/2025]
Abstract
The production of energy is one of the most fundamental requirements for organismal survival. Decreasing expression of Drosophila 9G8 , an mRNA splicing protein, specifically in adipose tissue results in triglyceride accumulation. Decreasing 9G8 in adipose also results in upregulation of the acyl-CoA synthetases Acsx4 and Acsx5 ; however, the functions of these genes in regulating lipid metabolism is not fully understood. Here, we decreased Acsx4 and Acsx5 in fly adipose tissue and this resulted in high triglycerides. This suggests that these genes regulate lipid breakdown, and their upregulation is perhaps compensating for the triglyceride accumulation observed when 9G8 levels are decreased.
Collapse
|
17
|
Zheng ZG, Zhang YP, Zhang XY, Qin MY, Xu YY, Wu H, Liu RQ, Wu QY, Wang MS, Zhang C, Zheng YQ, Dai JY, Li P, Yang H. Ergosterol alleviates hepatic steatosis and insulin resistance via promoting fatty acid β-oxidation by activating mitochondrial ACSL1. Cell Rep 2025; 44:115203. [PMID: 39799570 DOI: 10.1016/j.celrep.2024.115203] [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: 06/20/2024] [Revised: 11/22/2024] [Accepted: 12/23/2024] [Indexed: 01/15/2025] Open
Abstract
Sterols target sterol-sensing domain (SSD) proteins to lower cholesterol and circulating and hepatic triglyceride levels, but the mechanism remains unclear. In this study, we identify acyl-coenzyme A (CoA) synthetase long-chain family member 1 (ACSL1) as a direct target of ergosterol (ES). The C-terminal domain of ACSL1 undergoes conformational changes from closed to open, and ES may target the drug-binding pocket in the acetyl-CoA synthetase-like domain 1 (ASLD1) of ACSL1 to stabilize the closed conformation and maintain its activity. Moreover, ES is mainly enriched in the mitochondria and promotes fatty acid β-oxidation through ACSL1 allosteric activation. Structure-activity relationship analysis reveals how different structural sterols interact with the sterol-sensing domain-containing protein (SCAP) and ACSL1, explaining their regulatory effects on lipid metabolism. Moreover, our findings reveal that the combination of SCAP inhibitor 25-hydroxycholesterol (25-HC) and ES has a stronger lipid-lowering effect than alone.
Collapse
Affiliation(s)
- Zu-Guo Zheng
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China.
| | - Yi-Ping Zhang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xiao-Yu Zhang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Meng-Yao Qin
- Collaborative Innovation Center for Northwestern Chinese Medicine & School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Yin-Yue Xu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - He Wu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Run-Qing Liu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Qiu-Yi Wu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ming-Su Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Chong Zhang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yue-Qin Zheng
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jian-Ye Dai
- Collaborative Innovation Center for Northwestern Chinese Medicine & School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China.
| | - Ping Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China.
| | - Hua Yang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China.
| |
Collapse
|
18
|
Barnhart S, Shimizu-Albergine M, Kedar E, Kothari V, Shao B, Krueger M, Hsu CC, Tang J, Kanter JE, Kramer F, Djukovic D, Pascua V, Loo YM, Colonna L, Van den Bogaerde SJ, An J, Gale M, Reue K, Fisher EA, Gharib SA, Elkon KB, Bornfeldt KE. Type I IFN induces long-chain acyl-CoA synthetase 1 to generate a phosphatidic acid reservoir for lipotoxic saturated fatty acids. J Lipid Res 2025; 66:100730. [PMID: 39675509 PMCID: PMC11786746 DOI: 10.1016/j.jlr.2024.100730] [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: 09/23/2024] [Revised: 11/22/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024] Open
Abstract
Long-chain acyl-CoA synthetase 1 (ACSL1) catalyzes the conversion of long-chain fatty acids to acyl-CoAs. ACSL1 is required for β-oxidation in tissues that rely on fatty acids as fuel, but no consensus exists on why ACSL1 is induced by inflammatory mediators in immune cells. We used a comprehensive and unbiased approach to investigate the role of ACSL1 induction by interferon type I (IFN-I) in myeloid cells in vitro and in a mouse model of IFN-I overproduction. Our results show that IFN-I induces ACSL1 in macrophages via its interferon-α/β receptor, and consequently that expression of ACSL1 is increased in myeloid cells from individuals with systemic lupus erythematosus (SLE), an autoimmune condition characterized by increased IFN production. Taking advantage of a myeloid cell-targeted ACSL1-deficient mouse model and a series of lipidomics, proteomics, metabolomics and functional analyses, we show that IFN-I leverages induction of ACSL1 to increase accumulation of fully saturated phosphatidic acid species in macrophages. Conversely, ACSL1 induction is not needed for IFN-I's ability to induce the prototypical IFN-stimulated protein signature or to suppress proliferation or macrophage metabolism. Loss of ACSL1 in IFN-I stimulated myeloid cells enhances apoptosis and secondary necrosis in vitro, especially in the presence of increased saturated fatty acid load, and in a mouse model of atherosclerosis associated with IFN overproduction, resulting in larger lesion necrotic cores. We propose that ACSL1 induction is a mechanism used by IFN-I to increase phosphatidic acid saturation while protecting the cells from saturated fatty acid-induced cell death.
Collapse
Affiliation(s)
- Shelley Barnhart
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA; UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Masami Shimizu-Albergine
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA; UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Eyal Kedar
- Division of Rheumatology, University of Washington, Seattle, WA
| | - Vishal Kothari
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA; UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Baohai Shao
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA; UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Melissa Krueger
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA
| | - Cheng-Chieh Hsu
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA; UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Jingjing Tang
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA; UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Jenny E Kanter
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA; UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Farah Kramer
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA; UW Medicine Diabetes Institute, University of Washington, Seattle, WA
| | - Danijel Djukovic
- Department of Anesthesiology and Pain Medicine, Northwest Metabolomics Research Center, University of Washington, Seattle, WA
| | - Vadim Pascua
- Department of Anesthesiology and Pain Medicine, Northwest Metabolomics Research Center, University of Washington, Seattle, WA
| | - Yueh-Ming Loo
- Department of Immunology, University of Washington, Seattle, WA
| | | | | | - Jie An
- Division of Rheumatology, University of Washington, Seattle, WA
| | - Michael Gale
- Department of Immunology, University of Washington, Seattle, WA
| | - Karen Reue
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Edward A Fisher
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY
| | - Sina A Gharib
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA
| | - Keith B Elkon
- Division of Rheumatology, University of Washington, Seattle, WA
| | - Karin E Bornfeldt
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, Seattle, WA; UW Medicine Diabetes Institute, University of Washington, Seattle, WA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA.
| |
Collapse
|
19
|
Smith DM, Choi J, Wolfgang MJ. Tissue specific roles of fatty acid oxidation. Adv Biol Regul 2025; 95:101070. [PMID: 39672726 PMCID: PMC11832339 DOI: 10.1016/j.jbior.2024.101070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 12/03/2024] [Indexed: 12/15/2024]
Abstract
Mitochondrial long chain fatty acid β-oxidation is a critical central carbon catabolic process. The importance of fatty acid oxidation is made evident by the life-threatening disease associated with diverse inborn errors in the pathway. While inborn errors show multisystemic requirements for fatty acid oxidation, it is not clear from the clinical presentation of these enzyme deficiencies what the tissue specific roles of the pathway are compared to secondary systemic effects. To understand the cell or tissue specific contributions of fatty acid oxidation to systemic physiology, conditional knockouts in mice have been employed to determine the requirements of fatty acid oxidation in disparate cell types. This has produced a host of surprising results that sometimes run counter to the canonical view of this metabolic pathway. The rigor of conditional knockouts has also provided clarity over previous research utilizing cell lines in vitro or small molecule inhibitors with dubious specificity. Here we will summarize current research using mouse models of Carnitine Palmitoyltransferases to determine the tissue specific roles and requirements of long chain mitochondrial fatty acid β-oxidation.
Collapse
Affiliation(s)
- Danielle M Smith
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joseph Choi
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael J Wolfgang
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
20
|
Choi J, Smith DM, Scafidi S, Riddle RC, Wolfgang MJ. Carnitine palmitoyltransferase 1 facilitates fatty acid oxidation in a non-cell-autonomous manner. Cell Rep 2024; 43:115006. [PMID: 39671290 PMCID: PMC11726389 DOI: 10.1016/j.celrep.2024.115006] [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: 06/04/2024] [Revised: 09/18/2024] [Accepted: 11/06/2024] [Indexed: 12/15/2024] Open
Abstract
Mitochondrial fatty acid oxidation is facilitated by the combined activities of carnitine palmitoyltransferase 1 (Cpt1) and Cpt2, which generate and utilize acylcarnitines, respectively. We compare the response of mice with liver-specific deficiencies in the liver-enriched Cpt1a or the ubiquitously expressed Cpt2 and discover that they display unique metabolic, physiological, and molecular phenotypes. The loss of Cpt1a or Cpt2 results in the induction of the muscle-enriched isoenzyme Cpt1b in hepatocytes in a Pparα-dependent manner. However, hepatic Cpt1b does not contribute substantively to hepatic fatty acid oxidation when Cpt1a is absent. Liver-specific double knockout of Cpt1a and Cpt1b or Cpt2 eliminates the mitochondrial oxidation of non-esterified fatty acids. However, Cpt1a/Cpt1b double knockout mice retain fatty acid oxidation by utilizing extracellular long-chain acylcarnitines that are dependent on Cpt2. These data demonstrate the non-cell-autonomous intercellular metabolism of fatty acids in hepatocytes.
Collapse
Affiliation(s)
- Joseph Choi
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Danielle M Smith
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Susanna Scafidi
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ryan C Riddle
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, USA; Research and Development Service, Baltimore VA Medical Center, Baltimore, MD, USA
| | - Michael J Wolfgang
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
21
|
Gu W, Zeng B, Zhang Y, Zhao F, Lin X, Wang X, Liu N, Sun F, Zhou F, Zhang S, Dai Y. Acyl-CoA long-chain synthetase 1 (ACSL1) protects the endometrium from excess palmitic acid stress during decidualization. Cell Signal 2024; 124:111438. [PMID: 39343116 DOI: 10.1016/j.cellsig.2024.111438] [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: 08/12/2024] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Endometrial receptivity relies on the functional and morphological change of endometrium stromal cells (EnSCs) and epithelial cells in the secretory phase. Decidualization of ESCs and transitions in endometrium epithelial cells are crucial for successful uterine implantation and maintaining pregnancy. Accumulated data have demonstrated that decidualization is tightly coordinated by lipid metabolism. However, the lipidomic change and regulatory mechanism in uterine decidualization are still unknown. Our study showed that endometrium stromal cells and decidual stromal cells had different lipidomic profiles. Acyl-CoA long-chain synthetase 1 (ACSL1) which converts fatty acids to acyl-CoA expression was strongly elevated during decidualization. ACSL1 knockdown inhibited stromal-to-decidual cell transition and decreased the decidualization markers prolactin and Insulin-like growth factor-binding protein-1 (IGFBP1) expression through the AKT pathway. Lipid uptake was upregulated in stromal cells while lipid droplet accumulation was downregulated during decidualization. Meanwhile, silencing of ACSL1 led to impaired spare respiratory capacity, and downregulation of TFAM expression, indicating robust lipid metabolism. While palmitic acid addition impeded decidualization, overexpression of ACSL1 could partially reverse its effect. ACSL inhibitor Triacsin C significantly impeded decidualization in a three-dimensional coculture model consisting of endometrial stromal cells and epithelial cells. Knockdown of ACSL1 in stromal cells decreased the expression of the decidualization markers PAEP and SPP1 in epithelial cells. Collectively, ACSL1 is essential for uterine decidualization and protects stromal cells from excess palmitic acid stress.
Collapse
Affiliation(s)
- Weijia Gu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; Zhejiang Provincial Clinical Research Center for Reproductive Health and Disease, Hangzhou 310016, China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, China
| | - Biya Zeng
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; Zhejiang Provincial Clinical Research Center for Reproductive Health and Disease, Hangzhou 310016, China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, China
| | - Yi Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; Zhejiang Provincial Clinical Research Center for Reproductive Health and Disease, Hangzhou 310016, China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, China
| | - Fanxuan Zhao
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; Zhejiang Provincial Clinical Research Center for Reproductive Health and Disease, Hangzhou 310016, China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, China
| | - Xiang Lin
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; Zhejiang Provincial Clinical Research Center for Reproductive Health and Disease, Hangzhou 310016, China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, China
| | - Xinyu Wang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; Zhejiang Provincial Clinical Research Center for Reproductive Health and Disease, Hangzhou 310016, China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, China
| | - Na Liu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; Zhejiang Provincial Clinical Research Center for Reproductive Health and Disease, Hangzhou 310016, China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, China
| | - Fangying Sun
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; Zhejiang Provincial Clinical Research Center for Reproductive Health and Disease, Hangzhou 310016, China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, China
| | - Feng Zhou
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; Zhejiang Provincial Clinical Research Center for Reproductive Health and Disease, Hangzhou 310016, China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, China
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; Zhejiang Provincial Clinical Research Center for Reproductive Health and Disease, Hangzhou 310016, China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, China.
| | - Yongdong Dai
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China; Zhejiang Provincial Clinical Research Center for Reproductive Health and Disease, Hangzhou 310016, China; Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou 310016, China.
| |
Collapse
|
22
|
Ahmad F, Qaisar R. Nicotinamide riboside kinase 2: A unique target for skeletal muscle and cardiometabolic diseases. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167487. [PMID: 39216649 DOI: 10.1016/j.bbadis.2024.167487] [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: 06/05/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Myopathy leads to skeletal and cardiac muscle degeneration which is a major cause of physical disability and heart failure. Despite the therapeutic advancement the prevalence of particularly cardiac diseases is rising at an alarming rate and novel therapeutic targets are required. Nicotinamide riboside kinase-2 (NRK-2 or NMRK2) is a muscle-specific β1-integrin binding protein abundantly expressed in the skeletal muscle while only a trace amount is detected in the healthy cardiac muscle. The level in cardiac tissue is profoundly upregulated under pathogenic conditions such as ischemia and hypertension. NRK-2 was initially identified to regulate myoblast differentiation and to enhance the levels of NAD+, an important coenzyme that potentiates cellular energy production and stress resilience. Recent advancement has shown that NRK-2 critically regulates numerous cellular and molecular processes under pathogenic conditions to modulate the disease severity. Therefore, given its restricted expression in the cardiac and skeletal muscle, NRK-2 may serve as a unique therapeutic target. In this review, we provided a comprehensive overview of the diverse roles of NRK-2 played in different cardiac and muscular diseases and discussed the underlying molecular mechanisms in detail. Moreover, this review precisely examined how NRK-2 regulates metabolism in cardiac muscle, and how dysfunctional NRK-2 is associated with energetic deficit and impaired muscle function, manifesting various cardiac and skeletal muscle disease conditions.
Collapse
Affiliation(s)
- Firdos Ahmad
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Space Medicine Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Rizwan Qaisar
- Cardiovascular Research Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Space Medicine Group, Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
23
|
Jiang Y, Wang W, Wang H, Zhang X, Kong Y, Chen YQ, Zhu S. ACSL1 positively regulates adipogenic differentiation. Biochem Biophys Res Commun 2024; 735:150865. [PMID: 39442449 DOI: 10.1016/j.bbrc.2024.150865] [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: 10/13/2024] [Revised: 10/16/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Aberrant adipogenic differentiation is strongly associated with obesity and related metabolic diseases. Elucidating the key factors driving adipogenesis is an effective strategy for identifying novel therapeutic targets for treating metabolic diseases represented by obesity. In this study, transcriptomic techniques were employed to investigate the functional genes that regulate adipogenic differentiation in OP9 cells and 3T3-L1 cells. The findings indicated a notable upregulation of Acsl1 expression throughout the adipogenic differentiation process. Knocking down Acsl1 led to a decrease in the expression of genes associated with adipogenesis and a reduction in triglyceride accumulation. Additionally, Acsl1 overexpression promoted adipocyte differentiation and adipose-specific overexpression of Acsl1 markedly aggravated steatosis induced by a high-fat diet. Mechanistically, Cyp2f2, Dusp23 and Gstm2 are the crucial genes implicated in Acsl1-induced adipogenic differentiation. The findings of this study indicate that Acsl1 promotes adipogenesis and could serve as a potential therapeutic target for treating obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Yao Jiang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Wei Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China; The Second School of Clinical Medical, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hui Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Xiaoru Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Yuling Kong
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Yong Q Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China; MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, China.
| | - Shenglong Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China.
| |
Collapse
|
24
|
Bai X, Zhu Q, Combs M, Wabitsch M, Mack CP, Taylor JM. GRAF1 deficiency leads to defective brown adipose tissue differentiation and thermogenic response. Sci Rep 2024; 14:28692. [PMID: 39562682 PMCID: PMC11577055 DOI: 10.1038/s41598-024-79301-6] [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: 12/11/2023] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
Adipose tissue, which is crucial for the regulation of energy within the body, contains both white and brown adipocytes. White adipose tissue (WAT) primarily stores energy, while brown adipose tissue (BAT) plays a critical role in energy dissipation as heat, offering potential for therapies aimed at enhancing metabolic health. Regulation of the RhoA/ROCK pathway is crucial for appropriate specification, differentiation and maturation of both white and brown adipocytes. However, our knowledge of how this pathway is controlled within specific adipose depots remains unclear, and to date a RhoA regulator that selectively controls adipocyte browning has not been identified. Our study shows that GRAF1, a RhoGAP, is highly expressed in metabolically active tissues, and closely correlates with brown adipocyte differentiation in culture and in vivo. Mice with either global or adipocyte-specific GRAF1 deficiency exhibit impaired BAT maturation and compromised cold-induced thermogenesis. Moreover, defects in differentiation of human GRAF1-deficient brown preadipocytes can be rescued by treatment with a Rho kinase inhibitor. Collectively, these studies indicate that GRAF1 can selectively induce brown adipocyte differentiation and suggest that manipulating GRAF1 activity may hold promise for the future treatment of diseases related to metabolic dysfunction.
Collapse
Affiliation(s)
- Xue Bai
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Qiang Zhu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Matthew Combs
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Martin Wabitsch
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, 89075, Ulm, Germany
| | - Christopher P Mack
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
- McAllister Heart Institute, University of North Carolina, 160 North Medical Drive, 501 Brinkhous-Bullitt, CB# 7525, Chapel Hill, NC, 27599, USA
| | - Joan M Taylor
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA.
- McAllister Heart Institute, University of North Carolina, 160 North Medical Drive, 501 Brinkhous-Bullitt, CB# 7525, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
25
|
Desai A, Loureiro ZY, DeSouza T, Yang Q, Solivan-Rivera J, Corvera S. cAMP driven UCP1 induction in human adipocytes requires ATGL-catalyzed lipolysis. Mol Metab 2024; 90:102051. [PMID: 39454826 PMCID: PMC11585812 DOI: 10.1016/j.molmet.2024.102051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/08/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
OBJECTIVE The uncoupling protein 1 (UCP1) is induced in brown or "beige" adipocytes through catecholamine-induced cAMP signaling, which activates diverse transcription factors. UCP1 expression can also be enhanced by PPARγ agonists such as rosiglitazone (Rsg). However, it is unclear whether this upregulation results from de-novo differentiation of beige adipocytes from progenitor cells, or from the induction of UCP1 in pre-existing adipocytes. To explore this, we employed human adipocytes differentiated from progenitor cells and examined their acute response to Rsg, to the adenylate-cyclase activator forskolin (Fsk), or to both simultaneously. METHODS Adipocytes generated from primary human progenitor cells were differentiated without exposure to PPARγ agonists, and treated for 3, 6 or 78 h to Fsk, to Rsg, or to both simultaneously. Bulk RNASeq, RNAScope, RT-PCR, CRISPR-Cas9 mediated knockout, oxygen consumption and western blotting were used to assess cellular responses. RESULTS UCP1 mRNA expression was induced within 3 h of exposure to either Rsg or Fsk, indicating that Rsg's effect is independent on additional adipocyte differentiation. Although Rsg and Fsk induced distinct overall transcriptional responses, both induced genes associated with calcium metabolism, lipid droplet assembly, and mitochondrial remodeling, denoting core features of human adipocyte beiging. Unexpectedly, we found that Fsk-induced UCP1 expression was reduced by approximately 80% following CRISPR-Cas9-mediated knockout of PNPLA2, the gene encoding the triglyceride lipase ATGL. As anticipated, ATGL knockout suppressed lipolysis; however, the associated suppression of UCP1 induction indicates that maximal cAMP-mediated UCP1 induction requires products of ATGL-catalyzed lipolysis. Supporting this, we observed that the reduction in Fsk-stimulated UCP1 induction caused by ATGL knockout was reversed by Rsg, implying that the role of lipolysis in this process is to generate natural PPARγ agonists. CONCLUSIONS UCP1 transcription is known to be stimulated by transcription factors activated downstream of cAMP-dependent protein kinases. Here we demonstrate that UCP1 transcription can also be acutely induced through PPARγ-activation. Moreover, both pathways are activated in human adipocytes in response to cAMP, synergistically inducing UCP1 expression. The stimulation of PPARγ in response to cAMP may result from the production of natural PPARγ activating ligands through ATGL-mediated lipolysis.
Collapse
Affiliation(s)
- Anand Desai
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Zinger Yang Loureiro
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA; Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Tiffany DeSouza
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Qin Yang
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA; Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Javier Solivan-Rivera
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA; Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Silvia Corvera
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA; Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA; Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
26
|
Qu J, Tian L, Zhang M, Sun B, Chen L. SGLT2 inhibitor canagliflozin reduces visceral adipose tissue in db/db mice by modulating AMPK/KLF4 signaling and regulating mitochondrial dynamics to induce browning. Mol Cell Endocrinol 2024; 592:112320. [PMID: 38964727 DOI: 10.1016/j.mce.2024.112320] [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: 03/25/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/06/2024]
Abstract
Obesity is characterized by excessive accumulation of adipose tissue (mainly visceral). The morphology and function of mitochondria are crucial for regulating adipose browning and weight loss. Research suggests that the SGLT2 inhibitor canagliflozin may induce weight loss through an unknown mechanism, particularly targeting visceral adipose tissue. While Krueppel-Like Factor 4 (KLF4) is known to be essential for energy metabolism and mitochondrial function, its specific impact on visceral adipose tissue remains unclear. We administered canagliflozin to db/db mice for 8 weeks, or exposed adipocytes to canagliflozin for 24 h. The expression levels of browning markers, mitochondrial dynamics, and KLF4 were assessed. Then we validated the function of KLF4 through overexpression in vivo and in vitro. Adenosine monophosphate-activated protein kinase (AMPK) agonists, inhibitors, and KLF4 si-RNA were employed to elucidate the relationship between AMPK and KLF4. The findings demonstrated that canagliflozin significantly decreased body weight in db/db mice and augmented cold-induced thermogenesis. Additionally, canagliflozin increased the expression of mitochondrial fusion-related factors while reducing the levels of fission markers in epididymal white adipose tissue. These consistent findings were mirrored in canagliflozin-treated adipocytes. Similarly, overexpression of KLF4 in both adipocytes and db/db mice yielded comparable results. In all, canagliflozin mitigates obesity in db/db mice by promoting the brown visceral adipocyte phenotype through enhanced mitochondrial fusion via AMPK/KLF4 signaling.
Collapse
Affiliation(s)
- Jingru Qu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People's Republic of China
| | - Lei Tian
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People's Republic of China
| | - Man Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People's Republic of China
| | - Bei Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People's Republic of China.
| | - Liming Chen
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, People's Republic of China.
| |
Collapse
|
27
|
Yang M, Ge J, Liu YL, Wang HY, Wang ZH, Li DP, He R, Xie YY, Deng HY, Peng XM, Wang WS, Liu JD, Zhu ZZ, Yu XF, Maretich P, Kajimura S, Pan RP, Chen Y. Sortilin-mediated translocation of mitochondrial ACSL1 impairs adipocyte thermogenesis and energy expenditure in male mice. Nat Commun 2024; 15:7746. [PMID: 39232011 PMCID: PMC11374900 DOI: 10.1038/s41467-024-52218-4] [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: 06/16/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024] Open
Abstract
Beige fat activation involves a fuel switch to fatty acid oxidation following chronic cold adaptation. Mitochondrial acyl-CoA synthetase long-chain family member 1 (ACSL1) localizes in the mitochondria and plays a key role in fatty acid oxidation; however, the regulatory mechanism of the subcellular localization remains poorly understood. Here, we identify an endosomal trafficking component sortilin (encoded by Sort1) in adipose tissues that shows dynamic expression during beige fat activation and facilitates the translocation of ACSL1 from the mitochondria to the endolysosomal pathway for degradation. Depletion of sortilin in adipocytes results in an increase of mitochondrial ACSL1 and the activation of AMPK/PGC1α signaling, thereby activating beige fat and preventing high-fat diet (HFD)-induced obesity and insulin resistance. Collectively, our findings indicate that sortilin controls adipose tissue fatty acid oxidation by substrate fuel selection during beige fat activation and provides a potential targeted approach for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Min Yang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Ge
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Lian Liu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huan-Yu Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi-Han Wang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan-Pei Li
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui He
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Yu Xie
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-Yan Deng
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue-Min Peng
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen-She Wang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Dai Liu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zeng-Zhe Zhu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue-Feng Yu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Pema Maretich
- Research Laboratory of Electronics and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Howard Hughes Medical Institute, Boston, MA, USA
| | - Ru-Ping Pan
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Nuclear Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Chen
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China.
| |
Collapse
|
28
|
Jin Z, Wang X. Traditional Chinese medicine and plant-derived natural products in regulating triglyceride metabolism: Mechanisms and therapeutic potential. Pharmacol Res 2024; 208:107387. [PMID: 39216839 DOI: 10.1016/j.phrs.2024.107387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
The incidence of cardiometabolic disease is increasing globally, with a trend toward younger age of onset. Among these, atherosclerotic cardiovascular disease is a leading cause of mortality worldwide. Despite the efficacy of traditional lipid-lowering drugs, such as statins, in reducing low-density lipoprotein cholesterol levels, a significant residual risk of cardiovascular events remains, which is closely related to unmet triglyceride (TG) targets. The clinical application of current TG-lowering Western medicines has certain limitations, necessitating alternative or complementary therapeutic strategies. Traditional Chinese medicine (TCM) and plant-derived natural products, known for their safety owing to their natural origins and diverse biological activities, offer promising avenues for TG regulation with potentially fewer side effects. This review systematically summarises the mechanisms of TG metabolism and subsequently reviews the regulatory effects of TCM and plant-derived natural products on TG metabolism, including the inhibition of TG synthesis (via endogenous and exogenous pathways), promotion of TG catabolism, regulation of fatty acid absorption and transport, enhancement of lipophagy, modulation of the gut microbiota, and other mechanisms. In conclusion, through a comprehensive analysis of recent studies, this review consolidates the multifaceted regulatory roles of TCM and plant-derived natural products in TG metabolism and elucidates their potential as safer, multi-target therapeutic agents in managing hypertriglyceridemia and mitigating cardiovascular risk, thereby providing a basis for new drug development.
Collapse
Affiliation(s)
- Zhou Jin
- Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaolong Wang
- Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
29
|
Yue L, Wang Y, Wang C, Niu S, Dong X, Guan Y, Chen S. Empagliflozin improves aortic injury in obese mice by regulating fatty acid metabolism. Open Med (Wars) 2024; 19:20241012. [PMID: 39176252 PMCID: PMC11340858 DOI: 10.1515/med-2024-1012] [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: 02/18/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 08/24/2024] Open
Abstract
Background Empagliflozin has been shown in clinical studies to lower the risk of adverse cardiovascular events. Using proteomics, the current study aims to determine whether empagliflozin reduces aortic alterations in obese mice and to investigate its molecular mechanism of action. Methods We constructed obese mice and then treated them with empagliflozin. Changes in the weight of the mice were recorded. Blood glucose and lipid levels were measured in each group of mice, and changes in pulse wave velocity and aortic structure were recorded. In addition, changes in aortic protein expression were detected by proteomics and analyzed bioinformatically. Results Proteomics results showed that 507 differentially expressed proteins (DEPs) were identified in the comparison of normal and obese mice, while 90 DEPs were identified in the comparison of obese and empagliflozin-treated mice. Examination of these three groups revealed that DEPs were largely associated with the digestion of unsaturated fats. Among them, empagliflozin significantly reduced the expression of fatty acid synthase (FASN), acyl-CoA desaturase 3 (SCD3), ACSL1. and ACSL5 in the aorta of obesity-induced mice, and there was a close relationship between the four. Conclusion Empagliflozin reduced the protein expression of FASN, SCD3, ACSL1, and ACSL5 in the aorta of obese mice and improved aortic fatty acid metabolism and reduced vascular stiffness for vasoprotective effects.
Collapse
Affiliation(s)
- Lin Yue
- Department of Endocrinology, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei, 050000, P.R. China
| | - Yue Wang
- Department of Ultrasonography, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei, 050000, P.R. China
| | - Cuiying Wang
- Department of Endocrinology, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei, 050000, P.R. China
| | - Shu Niu
- Department of Endocrinology, Shijiazhuang People’s Hospital, Shijiazhuang, Hebei, 050000, P.R. China
| | - Xihong Dong
- Department of Endocrinology, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei, 050000, P.R. China
| | - Yaqing Guan
- Department of Endocrinology, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei, 050000, P.R. China
| | - Shuchun Chen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, 050000, P.R. China
| |
Collapse
|
30
|
Peppicelli S, Kersikla T, Menegazzi G, Andreucci E, Ruzzolini J, Nediani C, Bianchini F, Calorini L. The critical role of glutamine and fatty acids in the metabolic reprogramming of anoikis-resistant melanoma cells. Front Pharmacol 2024; 15:1422281. [PMID: 39175551 PMCID: PMC11338773 DOI: 10.3389/fphar.2024.1422281] [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: 04/23/2024] [Accepted: 07/25/2024] [Indexed: 08/24/2024] Open
Abstract
Introduction: Circulating tumor cells (CTCs) represent the sub-population of cells shed into the vasculature and able to survive in the bloodstream, adhere to target vascular endothelial cells, and re-growth into the distant organ. CTCs have been found in the blood of most solid tumor-bearing patients and are used as a diagnostic marker. Although a complex genotypic and phenotypic signature characterizes CTCs, the ability to survive in suspension constitutes the most critical property, known as resistance to anoikis, e.g., the ability to resist apoptosis resulting from a loss of substrate adhesion. Here, we selected melanoma cells resistant to anoikis, and we studied their metabolic reprogramming, with the aim of identifying new metabolic targets of CTCs. Methods: Subpopulations of melanoma cells expressing a high anoikis-resistant phenotype were selected by three consecutive rocking exposures in suspension and studied for their phenotypic and metabolic characteristics. Moreover, we tested the efficacy of different metabolic inhibitors targeting glycolysis (2DG), LDHA (LDHA-in-3), the mitochondrial electron transport chain complex I (rotenone), glutaminase (BPTES), fatty acid transporter (SSO), fatty acid synthase (denifanstat), CPT1 (etomoxir), to inhibit cell survival and colony formation ability after 24 h of rocking condition. Results: Anoikis-resistant cells displayed higher ability to grow in suspension on agarose-covered dishes respect to control cells, and higher cell viability and colony formation capability after a further step in rocking condition. They showed also an epithelial-to-mesenchymal transition associated with high invasiveness and a stemness-like phenotype. Anoikis-resistant melanoma cells in suspension showed a metabolic reprogramming from a characteristic glycolytic metabolism toward a more oxidative metabolism based on the use of glutamine and fatty acids, while re-adhesion on the dishes reversed the metabolism to glycolysis. The treatment with metabolic inhibitors highlighted the effectiveness of rotenone, BPTES, SSO, and etomoxir in reducing the viability and the colony formation ability of cells capable of surviving in suspension, confirming the dependence of their metabolism on oxidative phosphorylation, using glutamine and fatty acids as the most important fuels. Discussion: This finding opens up new therapeutic strategies based on metabolic inhibitors of glutaminase and fatty acid oxidation for the treatment of CTCs and melanoma metastases.
Collapse
Affiliation(s)
- S. Peppicelli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | | | | | | | | | | | - F. Bianchini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | | |
Collapse
|
31
|
Zhang R, Chen S, Zhao F, Wang W, Liu D, Chen L, Bai T, Wu Z, Ji L, Zhang J. Sulforaphane enhanced muscle growth by promoting lipid oxidation through modulating key signaling pathways. Biosci Rep 2024; 44:BSR20240084. [PMID: 38868980 PMCID: PMC11224001 DOI: 10.1042/bsr20240084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/29/2024] [Accepted: 06/12/2024] [Indexed: 06/14/2024] Open
Abstract
Sulforaphane (SFN) has shown diverse effects on human health and diseases. SFN was administered daily to C57BL/6J mice at doses of 1 mg/kg (SFN1) and 3 mg/kg (SFN3) for 8 weeks. Both doses of SFN accelerated body weight increment. The cross-sectional area and diameter of Longissimus dorsi (LD) muscle fibers were enlarged in SFN3 group. Triglyceride (TG) and total cholesterol (TC) levels in LD muscle were decreased in SFN groups. RNA sequencing results revealed that 2455 and 2318 differentially expressed genes (DEGs) were found in SFN1 and SFN3 groups, respectively. Based on GO enrichment analysis, 754 and 911 enriched GO terms in the SFN1 and SFN3 groups, respectively. KEGG enrichment analysis shown that one KEGG pathway was enriched in the SFN1 group, while six KEGG pathways were enriched in the SFN3 group. The expressions of nine selected DEGs validated with qRT-PCR were in line with the RNA sequencing data. Furthermore, SFN treatment influenced lipid and protein metabolism related pathways including AMPK signaling, fatty acid metabolism signaling, cholesterol metabolism signalling, PPAR signaling, peroxisome signaling, TGFβ signaling, and mTOR signaling. In summary, SFN elevated muscle fibers size and reduced TG and TC content of in LD muscle by modulating protein and lipid metabolism-related signaling pathways.
Collapse
Affiliation(s)
- Rui Zhang
- Meat Processing Key Laboratory of Sichuan Province, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Suqin Chen
- Meat Processing Key Laboratory of Sichuan Province, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Feng Zhao
- Department of Oncology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei Wang
- Meat Processing Key Laboratory of Sichuan Province, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Dayu Liu
- Meat Processing Key Laboratory of Sichuan Province, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Lin Chen
- Meat Processing Key Laboratory of Sichuan Province, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Ting Bai
- Meat Processing Key Laboratory of Sichuan Province, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Zhoulin Wu
- Meat Processing Key Laboratory of Sichuan Province, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Lili Ji
- Meat Processing Key Laboratory of Sichuan Province, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| | - Jiamin Zhang
- Meat Processing Key Laboratory of Sichuan Province, College of Food and Biological Engineering, Chengdu University, Chengdu, China
| |
Collapse
|
32
|
Yang F, Fan J, Yang R, Cun Y. Integrative analysis of blood transcriptome profiles in small-cell lung cancer patients for identification of novel chemotherapy resistance-related biomarkers. Front Immunol 2024; 15:1338162. [PMID: 38957470 PMCID: PMC11217175 DOI: 10.3389/fimmu.2024.1338162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 06/03/2024] [Indexed: 07/04/2024] Open
Abstract
Introduction Chemoresistance constitutes a prevalent factor that significantly impacts thesurvival of patients undergoing treatment for smal-cell lung cancer (SCLC). Chemotherapy resistance in SCLC patients is generally classified as primary or acquired resistance, each governedby distinct mechanisms that remain inadequately researched. Methods In this study, we performed transcriptome screening of peripheral blood plasma obtainedfrom 17 patients before and after receiving combined etoposide and platinum treatment. We firs testimated pseudo-single-cell analysis using xCell and ESTIMATE and identified differentially expressed genes (DEGs), then performed network analysis to discover key hub genes involved in chemotherapy resistance. Results Our analysis showed a significant increase in class-switched memory B cell scores acrossboth chemotherapy resistance patterns, indicating their potential crucial role in mediatingresistance. Moreover, network analysis identifed PRICKLE3, TNFSFI0, ACSLl and EP300 as potential contributors to primary resistance, with SNWl, SENP2 and SMNDCl emerging assignificant factors in acquired resistance, providing valuable insights into chemotherapy resistancein SCLC. Discussion These findings offer valuable insights for understanding chemotherapy resistance and related gene signatures in SCLC, which could help further biological validation studies.
Collapse
Affiliation(s)
- Fang Yang
- Department of the Second Medical Oncology, Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jinhua Fan
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Runxiang Yang
- Department of the Second Medical Oncology, Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yupeng Cun
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
33
|
Wu Y, Zhang J, Wang W, Wu D, Kang Y, Fu L. MARK4 aggravates cardiac dysfunction in mice with STZ-induced diabetic cardiomyopathy by regulating ACSL4-mediated myocardial lipid metabolism. Sci Rep 2024; 14:12978. [PMID: 38839927 PMCID: PMC11153581 DOI: 10.1038/s41598-024-64006-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/04/2024] [Indexed: 06/07/2024] Open
Abstract
Diabetic cardiomyopathy is a specific type of cardiomyopathy. In DCM, glucose uptake and utilization are impaired due to insulin deficiency or resistance, and the heart relies more heavily on fatty acid oxidation for energy, resulting in myocardial lipid toxicity-related injury. MARK4 is a member of the AMPK-related kinase family, and improves ischaemic heart failure through microtubule detyrosination. However, the role of MARK4 in cardiac regulation of metabolism is unclear. In this study, after successful establishment of a diabetic cardiomyopathy model induced by streptozotocin and a high-fat diet, MARK4 expression was found to be significantly increased in STZ-induced DCM mice. After AAV9-shMARK4 was administered through the tail vein, decreased expression of MARK4 alleviated diabetic myocardial damage, reduced oxidative stress and apoptosis, and facilitated cardiomyocyte mitochondrial fusion, and promoted myocardial lipid oxidation metabolism. In addition, through the RNA-seq analysis of differentially expressed genes, we found that MARK4 deficiency promoted lipid decomposition and oxidative metabolism by downregulating the expression of ACSL4, thus reducing myocardial lipid accumulation in the STZ-induced DCM model.
Collapse
Affiliation(s)
- Yi Wu
- Laboratory of Cardiovascular Internal Medicine Department, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Jingqi Zhang
- Laboratory of Cardiovascular Internal Medicine Department, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Weiyi Wang
- Laboratory of Cardiovascular Internal Medicine Department, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Dongdong Wu
- The First Affiliated Hospital of Jinzhou Medical University, 157 Renmin Street, Guta District, Jinzhou, 121000, China
| | - Yang Kang
- Laboratory of Cardiovascular Internal Medicine Department, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Lu Fu
- Laboratory of Cardiovascular Internal Medicine Department, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
34
|
Zhou S, Taskintuna K, Hum J, Gulati J, Olaya S, Steinman J, Golestaneh N. PGC-1α repression dysregulates lipid metabolism and induces lipid droplet accumulation in retinal pigment epithelium. Cell Death Dis 2024; 15:385. [PMID: 38824126 PMCID: PMC11144268 DOI: 10.1038/s41419-024-06762-y] [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: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 06/03/2024]
Abstract
Drusen, the yellow deposits under the retina, are composed of lipids and proteins, and represent a hallmark of age-related macular degeneration (AMD). Lipid droplets are also reported in the retinal pigment epithelium (RPE) from AMD donor eyes. However, the mechanisms underlying these disease phenotypes remain elusive. Previously, we showed that Pgc-1α repression, combined with a high-fat diet (HFD), induce drastic AMD-like phenotypes in mice. We also reported increased PGC-1α acetylation and subsequent deactivation in the RPE derived from AMD donor eyes. Here, through a series of in vivo and in vitro experiments, we sought to investigate the molecular mechanisms by which PGC-1α repression could influence RPE and retinal function. We show that PGC-1α plays an important role in RPE and retinal lipid metabolism and function. In mice, repression of Pgc-1α alone induced RPE and retinal degeneration and drusen-like deposits. In vitro inhibition of PGC1A by CRISPR-Cas9 gene editing in human RPE (ARPE19- PGC1A KO) affected the expression of genes responsible for lipid metabolism, fatty acid β-oxidation (FAO), fatty acid transport, low-density lipoprotein (LDL) uptake, cholesterol esterification, cholesterol biosynthesis, and cholesterol efflux. Moreover, inhibition of PGC1A in RPE cells caused lipid droplet accumulation and lipid peroxidation. ARPE19-PGC1A KO cells also showed reduced mitochondrial biosynthesis, impaired mitochondrial dynamics and activity, reduced antioxidant enzymes, decreased mitochondrial membrane potential, loss of cardiolipin, and increased susceptibility to oxidative stress. Our data demonstrate the crucial role of PGC-1α in regulating lipid metabolism. They provide new insights into the mechanisms involved in lipid and drusen accumulation in the RPE and retina during aging and AMD, which may pave the way for developing novel therapeutic strategies targeting PGC-1α.
Collapse
Affiliation(s)
- Shuyan Zhou
- Department of Ophthalmology, Georgetown University Medical Center, Washington, DC, 20007, USA
| | - Kaan Taskintuna
- Department of Ophthalmology, Georgetown University Medical Center, Washington, DC, 20007, USA
| | - Jacob Hum
- Department of Ophthalmology, Georgetown University Medical Center, Washington, DC, 20007, USA
| | - Jasmine Gulati
- Department of Ophthalmology, Georgetown University Medical Center, Washington, DC, 20007, USA
| | - Stephanie Olaya
- Department of Ophthalmology, Georgetown University Medical Center, Washington, DC, 20007, USA
| | - Jeremy Steinman
- Department of Ophthalmology, Georgetown University Medical Center, Washington, DC, 20007, USA
| | - Nady Golestaneh
- Department of Ophthalmology, Georgetown University Medical Center, Washington, DC, 20007, USA.
- Department of Neurology, Georgetown University Medical Center, Washington, DC, 20007, USA.
- Department of Biochemistry and Molecular & Cellular Biology, Washington, DC, 20007, USA.
| |
Collapse
|
35
|
Bai Y, Li J, Wei Y, Chen Z, Liu Z, Guo D, Jia X, Niu Y, Shi B, Zhang X, Zhao Z, Hu J, Han X, Wang J, Liu X, Li S. Proteome Analysis Related to Unsaturated Fatty Acid Synthesis by Interfering with Bovine Adipocyte ACSL1 Gene. Antioxidants (Basel) 2024; 13:641. [PMID: 38929080 PMCID: PMC11200461 DOI: 10.3390/antiox13060641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Unsaturated fatty acids (UFAs) in beef play a vital role in promoting human health. Long-chain fatty acyl-CoA synthase 1 (ACSL1) is a crucial gene for UFA synthesis in bovine adipocytes. To investigate the protein expression profile during UFA synthesis, we performed a proteomic analysis of bovine adipocytes by RNA interference and non-interference with ACSL1 using label-free techniques. A total of 3558 proteins were identified in both the NC and si-treated groups, of which 1428 were differentially expressed proteins (DEPs; fold change ≥ 1.2 or ≤ 0.83 and p-value < 0.05). The enrichment analysis of the DEPs revealed signaling pathways related to UFA synthesis or metabolism, including cAMP, oxytocin, fatty acid degradation, glycerol metabolism, insulin, and the regulation of lipolysis in adipocytes (p-value < 0.05). Furthermore, based on the enrichment analysis of the DEPs, we screened 50 DEPs that potentially influence the synthesis of UFAs and constructed an interaction network. Moreover, by integrating our previously published transcriptome data, this study established a regulatory network involving differentially expressed long non-coding RNAs (DELs), highlighting 21 DEPs and 13 DELs as key genes involved in UFA synthesis. These findings present potential candidate genes for further investigation into the molecular mechanisms underlying UFA synthesis in bovines, thereby offering insights to enhance the quality of beef and contribute to consumer health in future studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Zhidong Zhao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.B.); (J.L.); (Y.W.); (Z.C.); (Z.L.); (D.G.); (X.J.); (Y.N.); (B.S.); (X.Z.); (X.H.); (J.W.); (X.L.); (S.L.)
| | - Jiang Hu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.B.); (J.L.); (Y.W.); (Z.C.); (Z.L.); (D.G.); (X.J.); (Y.N.); (B.S.); (X.Z.); (X.H.); (J.W.); (X.L.); (S.L.)
| | | | | | | | | |
Collapse
|
36
|
Tamas C, Tamas F, Kovecsi A, Cehan A, Balasa A. Metabolic Contrasts: Fatty Acid Oxidation and Ketone Bodies in Healthy Brains vs. Glioblastoma Multiforme. Int J Mol Sci 2024; 25:5482. [PMID: 38791520 PMCID: PMC11122426 DOI: 10.3390/ijms25105482] [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: 04/09/2024] [Revised: 05/09/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
The metabolism of glucose and lipids plays a crucial role in the normal homeostasis of the body. Although glucose is the main energy substrate, in its absence, lipid metabolism becomes the primary source of energy. The main means of fatty acid oxidation (FAO) takes place in the mitochondrial matrix through β-oxidation. Glioblastoma (GBM) is the most common form of primary malignant brain tumor (45.6%), with an incidence of 3.1 per 100,000. The metabolic changes found in GBM cells and in the surrounding microenvironment are associated with proliferation, migration, and resistance to treatment. Tumor cells show a remodeling of metabolism with the use of glycolysis at the expense of oxidative phosphorylation (OXPHOS), known as the Warburg effect. Specialized fatty acids (FAs) transporters such as FAT, FABP, or FATP from the tumor microenvironment are overexpressed in GBM and contribute to the absorption and storage of an increased amount of lipids that will provide sufficient energy used for tumor growth and invasion. This review provides an overview of the key enzymes, transporters, and main regulatory pathways of FAs and ketone bodies (KBs) in normal versus GBM cells, highlighting the need to develop new therapeutic strategies to improve treatment efficacy in patients with GBM.
Collapse
Affiliation(s)
- Corina Tamas
- Doctoral School of Medicine and Pharmacy, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania;
- Department of Neurosurgery, Emergency Clinical County Hospital, 540136 Targu Mures, Romania;
- Department of Neurosurgery, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania
| | - Flaviu Tamas
- Doctoral School of Medicine and Pharmacy, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania;
- Department of Neurosurgery, Emergency Clinical County Hospital, 540136 Targu Mures, Romania;
- Department of Neurosurgery, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania
| | - Attila Kovecsi
- Department of Morphopathology, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania;
- Department of Morphopathology, Emergency Clinical County Hospital, 540136 Targu Mures, Romania
| | - Alina Cehan
- Department of Plastic, Esthetics and Reconstructive Surgery, Emergency Clinical County Hospital, 540136 Targu Mures, Romania;
| | - Adrian Balasa
- Department of Neurosurgery, Emergency Clinical County Hospital, 540136 Targu Mures, Romania;
- Department of Neurosurgery, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania
| |
Collapse
|
37
|
Safi R, Menéndez P, Pol A. Lipid droplets provide metabolic flexibility for cancer progression. FEBS Lett 2024; 598:1301-1327. [PMID: 38325881 DOI: 10.1002/1873-3468.14820] [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: 09/04/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
A hallmark of cancer cells is their remarkable ability to efficiently adapt to favorable and hostile environments. Due to a unique metabolic flexibility, tumor cells can grow even in the absence of extracellular nutrients or in stressful scenarios. To achieve this, cancer cells need large amounts of lipids to build membranes, synthesize lipid-derived molecules, and generate metabolic energy in the absence of other nutrients. Tumor cells potentiate strategies to obtain lipids from other cells, metabolic pathways to synthesize new lipids, and mechanisms for efficient storage, mobilization, and utilization of these lipids. Lipid droplets (LDs) are the organelles that collect and supply lipids in eukaryotes and it is increasingly recognized that the accumulation of LDs is a new hallmark of cancer cells. Furthermore, an active role of LD proteins in processes underlying tumorigenesis has been proposed. Here, by focusing on three major classes of LD-resident proteins (perilipins, lipases, and acyl-CoA synthetases), we provide an overview of the contribution of LDs to cancer progression and discuss the role of LD proteins during the proliferation, invasion, metastasis, apoptosis, and stemness of cancer cells.
Collapse
Affiliation(s)
- Rémi Safi
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Pablo Menéndez
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Spain
- Consorcio Investigación Biomédica en Red de Cancer, CIBER-ONC, ISCIII, Barcelona, Spain
- Spanish Network for Advanced Cell Therapies (TERAV), Barcelona, Spain
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Spain
| |
Collapse
|
38
|
Das S, Subramaniyam N, Alén R, Komakula SSB, Song Z, Ge X, Han H, Desert R, Athavale D, Magdaleno F, Chen W, Barahona I, Lantvit D, Guzman G, Nieto N. Ablation of secreted phosphoprotein-1 in hepatocytes increases fatty acid oxidation and ameliorates alcohol-associated liver disease. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:781-794. [PMID: 38503560 DOI: 10.1111/acer.15304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/30/2024] [Accepted: 03/04/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND Previously, we demonstrated that Spp1-/- mice exhibit a greater susceptibility to alcohol-induced liver injury than wild-type (WT) mice. Notably, alcohol triggers the expression of osteopontin (encoded by SPP1) in hepatocytes. However, the specific role of hepatocyte-derived SPP1 in either mitigating or exacerbating alcohol-associated liver disease (AALD) has yet to be elucidated. We hypothesized that hepatocyte-derived SPP1 plays a role in AALD by modulating the regulation of steatosis. METHODS We analyzed hepatic SPP1 expression using four publicly available datasets from patients with alcoholic hepatitis (AH). Additionally, we examined SPP1 expression in the livers of WT mice subjected to either a control or ethanol Lieber-DeCarli (LDC) diet for 6 weeks. We compared the relationship between SPP1 expression and significantly dysregulated genes in AH with controls using correlation and enrichment analyses. To investigate the specific impact of hepatocyte-derived SPP1, we generated hepatocyte-specific Spp1 knock-out (Spp1ΔHep) mice and subjected them to either a control or ethanol Lieber-DeCarli diet for 6 weeks. RESULTS Alcohol induced hepatic SPP1 expression in both humans and mice. Our analysis, focusing on genes correlated with SPP1, revealed an enrichment of fatty acid oxidation (FAO) in three datasets, and peroxisome proliferator-activated receptor signaling in one dataset. Notably, FAO genes correlating with SPP1 were downregulated in patients with AH. Ethanol-fed WT mice exhibited higher serum-free fatty acids (FFAs), adipose tissue lipolysis, and hepatic fatty acid (FA) transporters. In contrast, ethanol-fed Spp1ΔHep mice displayed lower liver triglycerides, FFAs, and serum alanine transaminase and greater FAO gene expression than WT mice, indicating a protective effect against AALD. Primary hepatocytes from Spp1∆Hep mice exhibited heightened expression of genes encoding proteins involved in FAO. CONCLUSIONS Alcohol induces the expression of SPP1 in hepatocytes, leading to impaired FAO and contributing to the development of AALD.
Collapse
Affiliation(s)
- Sukanta Das
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | - Rosa Alén
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | - Zhuolun Song
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Xiaodong Ge
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Hui Han
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Romain Desert
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Dipti Athavale
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Fernando Magdaleno
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Wei Chen
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Ines Barahona
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Daniel Lantvit
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Grace Guzman
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Natalia Nieto
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
- Research and Development Service, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| |
Collapse
|
39
|
Saukko-Paavola AJ, Klemm RW. Remodelling of mitochondrial function by import of specific lipids at multiple membrane-contact sites. FEBS Lett 2024; 598:1274-1291. [PMID: 38311340 DOI: 10.1002/1873-3468.14813] [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: 11/29/2023] [Revised: 12/14/2023] [Accepted: 12/28/2023] [Indexed: 02/08/2024]
Abstract
Organelles form physical and functional contact between each other to exchange information, metabolic intermediates, and signaling molecules. Tethering factors and contact site complexes bring partnering organelles into close spatial proximity to establish membrane contact sites (MCSs), which specialize in unique functions like lipid transport or Ca2+ signaling. Here, we discuss how MCSs form dynamic platforms that are important for lipid metabolism. We provide a perspective on how import of specific lipids from the ER and other organelles may contribute to remodeling of mitochondria during nutrient starvation. We speculate that mitochondrial adaptation is achieved by connecting several compartments into a highly dynamic organelle network. The lipid droplet appears to be a central hub in coordinating the function of these organelle neighborhoods.
Collapse
Affiliation(s)
| | - Robin W Klemm
- Department of Physiology, Anatomy and Genetics, University of Oxford, UK
| |
Collapse
|
40
|
Sharma AK, Khandelwal R, Wolfrum C. Futile lipid cycling: from biochemistry to physiology. Nat Metab 2024; 6:808-824. [PMID: 38459186 DOI: 10.1038/s42255-024-01003-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/02/2024] [Indexed: 03/10/2024]
Abstract
In the healthy state, the fat stored in our body isn't just inert. Rather, it is dynamically mobilized to maintain an adequate concentration of fatty acids (FAs) in our bloodstream. Our body tends to produce excess FAs to ensure that the FA availability is not limiting. The surplus FAs are actively re-esterified into glycerides, initiating a cycle of breakdown and resynthesis of glycerides. This cycle consumes energy without generating a new product and is commonly referred to as the 'futile lipid cycle' or the glyceride/FA cycle. Contrary to the notion that it's a wasteful process, it turns out this cycle is crucial for systemic metabolic homeostasis. It acts as a control point in intra-adipocyte and inter-organ cross-talk, a metabolic rheostat, an energy sensor and a lipid diversifying mechanism. In this Review, we discuss the metabolic regulation and physiological implications of the glyceride/FA cycle and its mechanistic underpinnings.
Collapse
Affiliation(s)
- Anand Kumar Sharma
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| | - Radhika Khandelwal
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Christian Wolfrum
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|
41
|
He W, Liu X, Feng Y, Ding H, Sun H, Li Z, Shi B. Dietary fat supplementation relieves cold temperature-induced energy stress through AMPK-mediated mitochondrial homeostasis in pigs. J Anim Sci Biotechnol 2024; 15:56. [PMID: 38584279 PMCID: PMC11000307 DOI: 10.1186/s40104-024-01014-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/14/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND Cold stress has negative effects on the growth and health of mammals, and has become a factor restricting livestock development at high latitudes and on plateaus. The gut-liver axis is central to energy metabolism, and the mechanisms by which it regulates host energy metabolism at cold temperatures have rarely been illustrated. In this study, we evaluated the status of glycolipid metabolism and oxidative stress in pigs based on the gut-liver axis and propose that AMP-activated protein kinase (AMPK) is a key target for alleviating energy stress at cold temperatures by dietary fat supplementation. RESULTS Dietary fat supplementation alleviated the negative effects of cold temperatures on growth performance and digestive enzymes, while hormonal homeostasis was also restored. Moreover, cold temperature exposure increased glucose transport in the jejunum. In contrast, we observed abnormalities in lipid metabolism, which was characterized by the accumulation of bile acids in the ileum and plasma. In addition, the results of the ileal metabolomic analysis were consistent with the energy metabolism measurements in the jejunum, and dietary fat supplementation increased the activity of the mitochondrial respiratory chain and lipid metabolism. As the central nexus of energy metabolism, the state of glycolipid metabolism and oxidative stress in the liver are inconsistent with that in the small intestine. Specifically, we found that cold temperature exposure increased glucose transport in the liver, which fully validates the idea that hormones can act on the liver to regulate glucose output. Additionally, dietary fat supplementation inhibited glucose transport and glycolysis, but increased gluconeogenesis, bile acid cycling, and lipid metabolism. Sustained activation of AMPK, which an energy receptor and regulator, leads to oxidative stress and apoptosis in the liver; dietary fat supplementation alleviates energy stress by reducing AMPK phosphorylation. CONCLUSIONS Cold stress reduced the growth performance and aggravated glycolipid metabolism disorders and oxidative stress damage in pigs. Dietary fat supplementation improved growth performance and alleviated cold temperature-induced energy stress through AMPK-mediated mitochondrial homeostasis. In this study, we highlight the importance of AMPK in dietary fat supplementation-mediated alleviation of host energy stress in response to environmental changes.
Collapse
Affiliation(s)
- Wei He
- College of Animal Science and Technology, Northeast Agricultural University, 600 Changjiang Street, Harbin, 150030, PR China
| | - Xinyu Liu
- College of Animal Science and Technology, Northeast Agricultural University, 600 Changjiang Street, Harbin, 150030, PR China
| | - Ye Feng
- College of Animal Science and Technology, Northeast Agricultural University, 600 Changjiang Street, Harbin, 150030, PR China
| | - Hongwei Ding
- College of Animal Science and Technology, Northeast Agricultural University, 600 Changjiang Street, Harbin, 150030, PR China
| | - Haoyang Sun
- College of Animal Science and Technology, Northeast Agricultural University, 600 Changjiang Street, Harbin, 150030, PR China
| | - Zhongyu Li
- College of Animal Science and Technology, Northeast Agricultural University, 600 Changjiang Street, Harbin, 150030, PR China
| | - Baoming Shi
- College of Animal Science and Technology, Northeast Agricultural University, 600 Changjiang Street, Harbin, 150030, PR China.
| |
Collapse
|
42
|
Li J, Dong Y, Zhou T, Tian H, Huang X, Zhang YQ, Wang Y, Lam SM, Shui G. Long-chain acyl-CoA synthetase regulates systemic lipid homeostasis via glycosylation-dependent lipoprotein production. LIFE METABOLISM 2024; 3:loae004. [PMID: 39872215 PMCID: PMC11749247 DOI: 10.1093/lifemeta/loae004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 01/04/2024] [Accepted: 01/16/2024] [Indexed: 01/30/2025]
Abstract
Interorgan lipid transport is crucial for organism development and the maintenance of physiological function. Here, we demonstrate that Drosophila long-chain acyl-CoA synthetase (dAcsl), which catalyzes the conversion of fatty acids into acyl-coenzyme As (acyl-CoAs), plays a critical role in regulating systemic lipid homeostasis. dAcsl deficiency in the fat body led to the ectopic accumulation of neutral lipids in the gut, along with significantly reduced lipoprotein contents in both the fat body and hemolymph. The aberrant phenotypes were rescued by fat body-specific overexpression of apolipophorin. A multi-omics investigation comprising lipidomics, metabolomics, and proteomics in conjunction with genetic screening revealed that glycosylation processes were suppressed in dAcsl knockdown flies. Overexpression of CG9035, human ortholog of which is implicated in the congenital disorder of glycosylation, ameliorated gut lipid accumulation in Drosophila. Aberrant lipoprotein glycosylation led to accelerated proteasome-related degradation and induced ER stress in dAcsl knockdown flies, impairing lipoprotein release into the circulation which compromised interorgan lipid transport between the fat body and the gut. Inhibition of ubiquitin-proteasome-dependent degradation alleviated the phenotype of gut ectopic fat accumulation in dAcsl knockdown flies. Finally, we verified that ACSL4, the human homolog of dAcsl, also regulated lipoprotein levels in HepG2 cells, indicating that the role of dAcsl in modulating lipoprotein secretion and systemic lipid homeostasis is possibly conserved in humans.
Collapse
Affiliation(s)
- Jie Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences , Beijing 100101, China
| | - Yue Dong
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences , Beijing 100101, China
| | - Tianxing Zhou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences , Beijing 100101, China
| | - He Tian
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiahe Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yong Q Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences , Beijing 100101, China
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences , Beijing 100101, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- Lipidall Technologies Company Limited, Changzhou, Jiangsu 213000, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences , Beijing 100101, China
| |
Collapse
|
43
|
You S, Lei G, Zhou H, Li J, Chen S, Huang J, Vasseur L, Gurr GM, You M, Chen Y. Thermal acclimation uncovers a simple genetic basis of adaptation to high temperature in a cosmopolitan pest. iScience 2024; 27:109242. [PMID: 38425842 PMCID: PMC10904271 DOI: 10.1016/j.isci.2024.109242] [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: 07/03/2023] [Revised: 10/16/2023] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
Understanding a population's fitness heterogeneity and genetic basis of thermal adaptation is essential for predicting the responses to global warming. We examined the thermotolerance and genetic adaptation of Plutella xylostella to exposure to hot temperatures. The population fitness parameters of the hot-acclimated DBM strains varied in the thermal environments. Using genome scanning and transcription profiling, we find a number of genes potentially involved in thermal adaptation of DBM. Editing two ABCG transporter genes, PxWhite and PxABCG, confirmed their role in altering cuticle permeability and influencing thermal responses. Our results demonstrate that SNP mutations in genes and changes in gene expression can allow DBM to rapidly adapt to thermal environment. ABCG transporter genes play an important role in thermal adaptation of DBM. This work improves our understanding of genetic adaptation mechanisms of insects to thermal stress and our capacity to predict the effects of rising global temperatures on ectotherms.
Collapse
Affiliation(s)
- Shijun You
- State Key Laboratory of Ecological Pest Control for Fujian-Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Institute of Plant Protection, Fujian Academy of Agricultural Sciences, Fuzhou 350002, China
- Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fuzhou 350002, China
- Ministerial and Provincial Joint Innovation Centre for Safety Production of Cross-Strait Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Gaoke Lei
- State Key Laboratory of Ecological Pest Control for Fujian-Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Institute of Plant Protection, Fujian Academy of Agricultural Sciences, Fuzhou 350002, China
- Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fuzhou 350002, China
- Ministerial and Provincial Joint Innovation Centre for Safety Production of Cross-Strait Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Huiling Zhou
- State Key Laboratory of Ecological Pest Control for Fujian-Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Institute of Plant Protection, Fujian Academy of Agricultural Sciences, Fuzhou 350002, China
- Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fuzhou 350002, China
- Ministerial and Provincial Joint Innovation Centre for Safety Production of Cross-Strait Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jianyu Li
- State Key Laboratory of Ecological Pest Control for Fujian-Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Institute of Plant Protection, Fujian Academy of Agricultural Sciences, Fuzhou 350002, China
- Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fuzhou 350002, China
- Ministerial and Provincial Joint Innovation Centre for Safety Production of Cross-Strait Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Shaoping Chen
- State Key Laboratory of Ecological Pest Control for Fujian-Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Institute of Plant Protection, Fujian Academy of Agricultural Sciences, Fuzhou 350002, China
- Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fuzhou 350002, China
- Ministerial and Provincial Joint Innovation Centre for Safety Production of Cross-Strait Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jieling Huang
- State Key Laboratory of Ecological Pest Control for Fujian-Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Institute of Plant Protection, Fujian Academy of Agricultural Sciences, Fuzhou 350002, China
- Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fuzhou 350002, China
- Ministerial and Provincial Joint Innovation Centre for Safety Production of Cross-Strait Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Liette Vasseur
- State Key Laboratory of Ecological Pest Control for Fujian-Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Institute of Plant Protection, Fujian Academy of Agricultural Sciences, Fuzhou 350002, China
- Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fuzhou 350002, China
- Department of Biological Sciences, UNESCO Chair on Community Sustainability, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Geoff M. Gurr
- State Key Laboratory of Ecological Pest Control for Fujian-Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Institute of Plant Protection, Fujian Academy of Agricultural Sciences, Fuzhou 350002, China
- Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fuzhou 350002, China
- Gulbali Institute, Charles Sturt University, Orange, NSW 2800, Australia
| | - Minsheng You
- State Key Laboratory of Ecological Pest Control for Fujian-Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Institute of Plant Protection, Fujian Academy of Agricultural Sciences, Fuzhou 350002, China
- Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fuzhou 350002, China
- Ministerial and Provincial Joint Innovation Centre for Safety Production of Cross-Strait Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yanting Chen
- State Key Laboratory of Ecological Pest Control for Fujian-Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Institute of Plant Protection, Fujian Academy of Agricultural Sciences, Fuzhou 350002, China
- Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fuzhou 350002, China
- Ministerial and Provincial Joint Innovation Centre for Safety Production of Cross-Strait Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| |
Collapse
|
44
|
Su M, Chen F, Han D, Song M, Wang Y. PRMT7-Dependent Transcriptional Activation of Hmgb2 Aggravates Severe Acute Pancreatitis by Promoting Acsl1-Induced Ferroptosis. J Proteome Res 2024; 23:1075-1087. [PMID: 38376246 DOI: 10.1021/acs.jproteome.3c00830] [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] [Indexed: 02/21/2024]
Abstract
Severe acute pancreatitis (SAP) is a highly fatal abdominal emergency, and its association with protein arginine methyltransferase 7 (PRMT7), the sole known type III enzyme responsible for the monomethylation of arginine residue, remains unexplored. In this study, we observe an increase in the PRMT7 levels in the pancreas of SAP mice and Cerulein-LPS-stimulated AR42J cells. Overexpression of Prmt7 exacerbated pancreatic damage in SAP, while the inhibition of PRMT7 improved SAP-induced pancreatic damage. Furthermore, PRMT7 overexpression promoted inflammation, oxidative stress, and ferroptosis during SAP. Mechanically, PRMT7 catalyzed monomethylation at histone H4 arginine 3 (H4R3me1) at the promoter region of high mobility group proteins 2 (HMGB2), thereby enhancing its transcriptional activity. Subsequently, HMGB2 facilitated Acyl CoA synthase long-chain family member 1 (ACSL1) transcription by binding to its promoter region, resulting in the activation of ferroptosis. Inhibition of PRMT7 effectively alleviated ferroptosis in Cerulein-LPS-induced AR42J cells by suppressing the HMGB2-ACSL1 pathway. Overall, our study reveals that PRMT7 plays a crucial role in promoting SAP through its regulation of the HMGB2-ACSL1 pathway to accelerate ferroptosis.
Collapse
Affiliation(s)
- Minghua Su
- Department of Emergency Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China
| | - Feng Chen
- Department of Emergency Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China
| | - Dong Han
- Department of Emergency Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China
| | - Menglong Song
- Emergency Intensive Care Unit, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China
| | - Yifan Wang
- Department of Emergency Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China
| |
Collapse
|
45
|
Wang W, Wang P, Zhu L, Liu B, Wei Q, Hou Y, Li X, Hu Y, Li W, Wang Y, Jiang C, Yang G, Wang J. An optimized fluorescent biosensor for monitoring long-chain fatty acyl-CoAs metabolism in vivo. Biosens Bioelectron 2024; 247:115935. [PMID: 38128319 DOI: 10.1016/j.bios.2023.115935] [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: 10/17/2023] [Revised: 12/07/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
Long-chain fatty acyl-CoAs (LCACoAs) are intermediates in lipid metabolism that exert a wide range of cellular functions. However, our knowledge about the subcellular distribution and regulatory impacts of LCACoAs is limited by a lack of methods for detecting LCACoAs in living cells and tissues. Here, we report our development of LACSerHR, a genetically encoded fluorescent biosensor that enables precise measurement of subtle fluctuations in the levels of endogenous LCACoAs in vivo. LACSerHR significantly improve the fluorescent brightness and analyte affinity, in vitro and in vivo testing showcased LACSerHR's large dynamic range. We demonstrate LACSerHR's capacity for real-time evaluation of LCACoA levels in specific subcellular compartments, for example in response to disruption of ACSL enzyme function in HEK293T cells. Moreover, we show the application of LACSerHR for sensitive measurement of elevated LCACoA levels in the livers of mouse models for two common metabolic diseases (NAFLD and type 2 diabetes). Thus, our LACSerHR sensor is a powerful, broadly applicable tool for studying LCACoAs metabolism and disease.
Collapse
Affiliation(s)
- Weibo Wang
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China; National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan, 430079, PR China
| | - Pengcheng Wang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, PR China
| | - Lixin Zhu
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Bingjie Liu
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Qingpeng Wei
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Yongkang Hou
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Xi Li
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Yufei Hu
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Wenzhe Li
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Yuan Wang
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, PR China
| | - Guangfu Yang
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan, 430079, PR China.
| | - Jing Wang
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China.
| |
Collapse
|
46
|
Zhong Q, Wang X, Wei R, Liu F, Alamin M, Sun J, Gui L. Equisetin inhibits adiposity through AMPK-dependent regulation of brown adipocyte differentiation. Heliyon 2024; 10:e25458. [PMID: 38327434 PMCID: PMC10847917 DOI: 10.1016/j.heliyon.2024.e25458] [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: 11/15/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/09/2024] Open
Abstract
Obesity has a significant impact on endocrine function, which leads to metabolic diseases including diabetes, insulin resistance, and other complications associated with obesity. Development of effective and safe anti-obesity drugs is imperative and necessary. Equisetin (EQST), a tetramate-containing marine fungal product, was reported to inhibit bacterial fatty acid synthesis and affect mitochondrial metabolism. It is tempting to speculate that EQST might have anti-obesity effects. This study was designed to explore anti-obesity effects and underlying mechanism of EQST on 3T3-L1 adipocytes differentiated from 3T3-L1 cells. Oil Red O staining showed that EQST reduced lipid accumulation in 3T3-L1 adipocytes. Quantitative real-time polymerase chain reaction and Western blot analysis revealed that EQST significantly inhibited expression of adipogenesis/lipogenesis-related genes C/ebp-α, Ppar-γ, Srebp1c, Fas, and reduced protein levels. There was also increased expression of key genes and protein levels involved in lipolysis (Perilipin, Atgl, Hsl), brown adipocyte differentiation (Prdm16, Ucp1), mitochondrial biogenesis (Pgc1α, Tfam) and β-oxidation Acsl1, Cpt1. Moreover, mitochondrial content, their membrane potential ΔΨM, and respiratory chain genes Mt-Co1, Cox7a1, Cox8b, and Cox4 (and protein) exhibited marked increase in expression upon EQST treatment, along with increased protein levels. Importantly, EQST induced expression and activation of AMPK, which was compromised by the AMPK inhibitor dorsomorphin, leading to rescue of EQST-downregulated Fas expression and a reduction of the EQST-increased expression of Pgc1α, Ucp1, and Cox4. Together, EQST robustly promotes fat clearance through the AMPK pathway, these results supporting EQST as a strong candidate for the development into an anti-obesity therapeutic agent.
Collapse
Affiliation(s)
- Qin Zhong
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, University Town, Gui'an New District, Guiyang City, Guizhou Province 550025, China
- Clinical Medical Research Center, Affiliated Hospital of Guizhou Medical University No.28 Beijing Road, Guiyang City, Guizhou Province 550001, China
| | - Xian Wang
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, University Town, Gui'an New District, Guiyang City, Guizhou Province 550025, China
| | - Ruiran Wei
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, University Town, Gui'an New District, Guiyang City, Guizhou Province 550025, China
- Department of Basic Medical Sciences, Clinical College of Anhui Medical University, No.69 Meishan Road Hefei City, Anhui Province 230031, China
| | - Fang Liu
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, University Town, Gui'an New District, Guiyang City, Guizhou Province 550025, China
| | - Md Alamin
- Department of Biology, College of Life Sciences, Southern Medical University of Science and Technology, No.1088 Xueyuan Road, Shenzhen City, Guangdong Province 518055, China
| | - Jiajia Sun
- Institute of Obstetrics and Gynecology, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, No.1120 Lianhua Road, Futian District, Shenzhen City, Guangdong Province 518000, China
| | - Liming Gui
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, University Town, Gui'an New District, Guiyang City, Guizhou Province 550025, China
- Institute of Obstetrics and Gynecology, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, No.1120 Lianhua Road, Futian District, Shenzhen City, Guangdong Province 518000, China
| |
Collapse
|
47
|
He W, Ding H, Feng Y, Liu X, Fang X, Gao F, Shi B. Dietary-fat supplementation alleviates cold temperature-induced metabolic dysbiosis and barrier impairment by remodeling gut microbiota. Food Funct 2024; 15:1443-1459. [PMID: 38226701 DOI: 10.1039/d3fo04916g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
As important components of the mammalian diet and tissues, fats are involved in a variety of biological processes in addition to providing energy. In general, the increase in basal metabolism and health risks under cold temperature conditions causes the host to need more energy to maintain body temperature and normal biological processes. The intestine and its microbiota are key components in orchestrating host metabolic homeostasis and immunity, and respond rapidly to changing environmental conditions. However, the role of dietary-fat supplementation in regulating host homeostasis of metabolism and barrier functions through gut microbiota at cold temperatures is incompletely understood. Our results showed that dietary-fat supplementation alleviated the negative effects of cold temperatures on the alpha-diversity of both ileal and colonic microbiota. Cold temperatures altered the ileal and colonic microbiota of pigs, and the extent of changes was more pronounced in the colonic microbiota. Translocation of the gut microbiota was restored after supplementation with a high-fat diet. In addition, cold temperatures exacerbated ileal mucosal damage and inflammation, and disrupted barrier function, which may be associated with decreased concentrations of butyrate and isobutyrate. Cold temperature-induced metabolic dysbiosis was manifested by altered hormone levels and upregulation of expression of multiple metabolites involved in metabolism (lipids, amino acids and minerals) and the immune response. Supplementation with a high-fat diet restored metabolic homeostasis and barrier function by improving gut-microbiota composition and increasing SCFAs concentrations in pigs. In conclusion, cold temperatures induced severe translocation of microbiota and barrier damage. These actions increased the risk of metabolic imbalance. Dietary-fat supplementation alleviated the adverse effects of cold temperatures on host metabolism by remodeling the gut microbiota.
Collapse
Affiliation(s)
- Wei He
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China.
| | - Hongwei Ding
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China.
| | - Ye Feng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China.
| | - Xinyu Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China.
| | - Xiuyu Fang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China.
| | - Feng Gao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China.
| | - Baoming Shi
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
48
|
Wang Y, Wang DY, Bu KN, Gao JD, Zhang BL. Prognosis prediction and risk stratification of breast cancer patients based on a mitochondria-related gene signature. Sci Rep 2024; 14:2859. [PMID: 38310106 PMCID: PMC10838276 DOI: 10.1038/s41598-024-52981-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 01/25/2024] [Indexed: 02/05/2024] Open
Abstract
As the malignancy with the highest global incidence, breast cancer represents a significant threat to women's health. Recent advances have shed light on the importance of mitochondrial function in cancer, particularly in metabolic reprogramming within tumors. Recognizing this, we developed a novel risk signature based on mitochondrial-related genes to improve prognosis prediction and risk stratification in breast cancer patients. In this study, transcriptome data and clinical features of breast cancer samples were extracted from two sources: the TCGA, serving as the training set, and the METABRIC, used as the independent validation set. We developed the signature using LASSO-Cox regression and assessed its prognostic efficacy via ROC curves. Furthermore, the signature was integrated with clinical features to create a Nomogram model, whose accuracy was validated through clinical calibration curves and decision curve analysis. To further elucidate prognostic variations between high and low-risk groups, we conducted functional enrichment and immune infiltration analyses. Additionally, the study encompassed a comparison of mutation landscapes and drug sensitivity, providing a comprehensive understanding of the differing characteristics in these groups. Conclusively, we established a risk signature comprising 8 mitochondrial-related genes-ACSL1, ALDH2, MTHFD2, MRPL13, TP53AIP1, SLC1A1, ME3, and BCL2A1. This signature was identified as an independent risk predictor for breast cancer patient survival, exhibiting a significant high hazard ratio (HR = 3.028, 95%CI 2.038-4.499, P < 0.001). Patients in the low-risk group showed a more favorable prognosis, with enhanced immune infiltration, distinct mutation landscapes, and greater sensitivity to anti-tumor drugs. In contrast, the high-risk group exhibited an adverse trend in these aspects. This risk signature represents a novel and effective prognostic indicator, suggesting valuable insights for patient stratification in breast cancer.
Collapse
Affiliation(s)
- Yang Wang
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ding-Yuan Wang
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ke-Na Bu
- Xingyuan Hospital of Yulin City, Yulin City, 719051, Shanxi Province, China
| | - Ji-Dong Gao
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union College, Shenzhen, 518116, China.
| | - Bai-Lin Zhang
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
49
|
Zhang J, Cheng X, Wei Y, Zhang Z, Zhou Q, Guan Y, Yan Y, Wang R, Jia C, An J, He M. Epigenome-wide perspective of cadmium-associated DNA methylation and its mediation role in the associations of cadmium with lipid levels and dyslipidemia risk. Food Chem Toxicol 2024; 184:114409. [PMID: 38128686 DOI: 10.1016/j.fct.2023.114409] [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: 05/16/2023] [Revised: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Studies demonstrated the associations of cadmium (Cd) with lipid levels and dyslipidemia risk, but the mechanisms involved need further exploration. OBJECTIVES We aimed to explore the role of DNA methylation (DNAM) in the relationship of Cd with lipid levels and dyslipidemia risk. METHODS Urinary cadmium levels (UCd) were measured by inductively coupled plasma mass spectrometry, serum high-density lipoprotein (HDL), total cholesterol, triglyceride, and low-density lipoprotein were measured with kits, and DNAM was measured using the Infinium MethylationEPIC BeadChip. Robust linear regressions were conducted for epigenome-wide association study. Multivariate linear and logistic regressions were performed to explore the associations of UCd with lipid levels and dyslipidemia risk, respectively. Mediation analyses were conducted to explore potential mediating role of DNAM in the associations of Cd with lipid levels and dyslipidemia risk. RESULTS UCd was negatively associated with HDL levels (p = 0.01) and positively associated with dyslipidemia (p < 0.01). There were 92/11 DMPs/DMRs (FDR<0.05) associated with UCd. Cd-associated DNAM and pathways were connected with cardiometabolic diseases and immunity. Cg07829377 (LINC01060) mediated 42.05%/22.88% of the UCd-HDL/UCd-dyslipidemia associations (p = 0.02 and 0.01, respectively). CONCLUSIONS Cadmium caused site-specific DNAM alterations and the associations of UCd with lipid levels and dyslipidemia risk may be partially mediated by DNAM.
Collapse
Affiliation(s)
- Jiazhen Zhang
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Xu Cheng
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yue Wei
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China; Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, 530021, China
| | - Zefang Zhang
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China; Department of Tuberculosis Control, Wuhan Pulmonary Hospital, Wuhan, Hubei, China
| | - Qihang Zhou
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Youbing Guan
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yan Yan
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Ruixin Wang
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chengyong Jia
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Jun An
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Meian He
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
50
|
Wang CH, Surbhi, Goraya S, Byun J, Pennathur S. Fatty acids and inflammatory stimuli induce expression of long-chain acyl-CoA synthetase 1 to promote lipid remodeling in diabetic kidney disease. J Biol Chem 2024; 300:105502. [PMID: 38016515 PMCID: PMC10770716 DOI: 10.1016/j.jbc.2023.105502] [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: 05/18/2023] [Revised: 10/30/2023] [Accepted: 11/10/2023] [Indexed: 11/30/2023] Open
Abstract
Fatty acid handling and complex lipid synthesis are altered in the kidney cortex of diabetic patients. We recently showed that inhibition of the renin-angiotensin system without changes in glycemia can reverse diabetic kidney disease (DKD) and restore the lipid metabolic network in the kidney cortex of diabetic (db/db) mice, raising the possibility that lipid remodeling may play a central role in DKD. However, the roles of specific enzymes involved in lipid remodeling in DKD have not been elucidated. In the present study, we used this diabetic mouse model and a proximal tubule epithelial cell line (HK2) to investigate the potential relationship between long-chain acyl-CoA synthetase 1 (ACSL1) and lipid metabolism in response to fatty acid exposure and inflammatory signals. We found ACSL1 expression was significantly increased in the kidney cortex of db/db mice, and exposure to palmitate or tumor necrosis factor-α significantly increased Acsl1 mRNA expression in HK-2 cells. In addition, palmitate treatment significantly increased the levels of long-chain acylcarnitines and fatty acyl CoAs in HK2 cells, and these increases were abolished in HK2 cell lines with specific deletion of Acsl1(Acsl1KO cells), suggesting a key role for ACSL1 in fatty acid β-oxidation. In contrast, tumor necrosis factor-α treatment significantly increased the levels of short-chain acylcarnitines and long-chain fatty acyl CoAs in HK2 cells but not in Acsl1KO cells, consistent with fatty acid channeling to complex lipids. Taken together, our data demonstrate a key role for ACSL1 in regulating lipid metabolism, fatty acid partitioning, and inflammation.
Collapse
Affiliation(s)
- Chih-Hong Wang
- Department of Physiology, Tulane University of School Medicine, New Orleans, Louisiana, USA; Tulane Hypertension & Renal Center of Excellence, Tulane University, New Orleans, Louisiana, USA; Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Surbhi
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Sayhaan Goraya
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jaeman Byun
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Subramaniam Pennathur
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|