1
|
Barbhuiya PA, Yoshitomi R, Pathak MP. Understanding the Link Between Sterol Regulatory Element Binding Protein (SREBPs) and Metabolic Dysfunction Associated Steatotic Liver Disease (MASLD). Curr Obes Rep 2025; 14:36. [PMID: 40227546 DOI: 10.1007/s13679-025-00626-y] [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] [Accepted: 04/01/2025] [Indexed: 04/15/2025]
Abstract
PURPOSE OF THE REVIEW This review aims to summarize the current scientific understanding on the complex interplay between sterol regulatory element-binding proteins (SREBPs) and metabolic dysfunction associated steatotic liver disease (MASLD) by critically examining a few significant molecular pathways. Additionally, the review explores the potential of both natural and synthetic SREBP inhibitors as promising therapeutic candidates for MASLD. RECENT FINDINGS SREBPs are central regulators of lipid homeostasis, with SREBP-1c primarily controlling fatty acid synthesis and SREBP-2 regulating cholesterol metabolism. Dysregulation of SREBP activity, often triggered by excessive caloric intake, insulin resistance, or endoplasmic reticulum (ER) stress, contributes to the development of metabolic syndrome and MASLD. SREBP-1c overexpression leads to increased de novo lipogenesis (DNL), hepatic lipid accumulation, and insulin resistance, while SREBP-2 modulates cholesterol metabolism via miRNA-33 and ABCA1 regulation leading to the pathogenesis of MASLD. The PI3K-Akt-mTORC1 pathway plays a critical role in SREBP activation, linking nutrient availability to lipid synthesis. Synthetic SREBP inhibitors, such as fatostatin and 25-hydroxycholesterol, and natural compounds, including kaempferol and resveratrol, show promise in modulating SREBP activity in vivo. CONCLUSION While targeting SREBP pathways presents a promising avenue for mitigating MASLD, further scientific investigation is imperative to identify and validate potential molecular targets. Although current studies on synthetic and natural SREBP inhibitors demonstrate encouraging results, rigorous pre-clinical and clinical research is warranted to translate these findings into effective MASLD treatments.
Collapse
Affiliation(s)
- Pervej Alom Barbhuiya
- Faculty of Pharmaceutical Science, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, 781026
- Centre for Research on Ethnomedicine, Faculty of Pharmaceutical Science, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, 781026
| | - Ren Yoshitomi
- National Institute of Advanced Industrial Science and Technology, AIST, Tokyo, Japan
| | - Manash Pratim Pathak
- Faculty of Pharmaceutical Science, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, 781026.
- Centre for Research on Ethnomedicine, Faculty of Pharmaceutical Science, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, 781026.
| |
Collapse
|
2
|
Guo XJ, Zhu BB, Li J, Guo P, Niu YB, Shi JL, Yokoyama W, Huang QS, Shao DY. Cholesterol metabolism in tumor immunity: Mechanisms and therapeutic opportunities for cancer. Biochem Pharmacol 2025; 234:116802. [PMID: 39954742 DOI: 10.1016/j.bcp.2025.116802] [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: 10/31/2024] [Revised: 12/25/2024] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Cholesterol is an essential component of the cell membrane which plays a critical role in the survival of immune and tumor cells. Reprogramming of cholesterol metabolism in both tumor cells and immune cells can impact tumor progression and anti-tumor immune responses. Strategies aimed at modulating cholesterol metabolism have been demonstrated to be effective in hindering tumor growth and boosting anti-tumor immune functions. This review provides a thorough analysis of intracellular cholesterol homeostasis regulation in cells, focusing on key genes and signaling pathways. It particularly emphasizes the regulatory mechanisms and importance of the cholesterol presence state (esterified/free), levels of cholesterol, and its metabolites in immune and tumor cells. Additionally, the review thoroughly explores how cholesterol metabolism and sources (endogenous/exogenous) in the tumor microenvironment (TME) contribute to the interplay among tumor cells, immune suppressor cells, and immune effector cells, promoting cancer progression and immune evasion. It also delves into current insights on the influence of cholesterol metabolites and related drugs in regulating tumor development or immunotherapy. Finally, it presents an overview of recent advancements in clinical and preclinical trials investigating the efficacy of targeted cholesterol metabolism treatments and combination therapies in cancer management, while proposing potential future research directions in tumor immunity. This review is poised to offer fresh perspectives and avenues for examining the potential of cancer immunotherapy centered on cholesterol metabolism regulation.
Collapse
Affiliation(s)
- Xiao-Jia Guo
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi 710072, PR China
| | - Bo-Bo Zhu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi 710072, PR China
| | - Jing Li
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an 710072, PR China
| | - Ping Guo
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi 710072, PR China
| | - Yin-Bo Niu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi 710072, PR China
| | - Jun-Ling Shi
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi 710072, PR China
| | - Wallace Yokoyama
- Processed Foods Research Unit, Western Regional Research Center, Agricultural Research Service, USDA, Albany, CA 94710, USA
| | - Qing-Sheng Huang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi 710072, PR China.
| | - Dong-Yan Shao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi West Road, Xi'an, Shaanxi 710072, PR China; Research & Development Institute of Northwestern Polytechnical University in Shenzhen, No. 45th, Gaoxin South 9th Road, Nanshan District, Shenzhen City 518063, PR China.
| |
Collapse
|
3
|
Wu Y, Song W, Su M, He J, Hu R, Zhao Y. The Role of Cholesterol Metabolism and Its Regulation in Tumor Development. Cancer Med 2025; 14:e70783. [PMID: 40145543 PMCID: PMC11948085 DOI: 10.1002/cam4.70783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/27/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Within the tumor microenvironment, tumor cells undergo metabolic reprogramming of cholesterol due to intrinsic cellular alterations and changes in the extracellular milieu. Furthermore, cholesterol reprogramming within this microenvironment influences the immune landscape of tumors, facilitating immune evasion and consequently promoting tumorigenesis. These biological changes involve modifications in numerous enzymes associated with cholesterol uptake and synthesis, including NPC1L1, SREBP, HMGCR, SQLE, and PCSK9. REVIEW This review systematically summarizes the role of cholesterol metabolism and its associated enzymes in cancer progression, examines the mechanisms through which dysregulation of cholesterol metabolism affects immune cells within the tumor microenvironment, and discusses recent advancements in cancer therapies that target cholesterol metabolism. CONCLUSION Targeting cholesterol metabolism-related enzymes can inhibit tumor growth, reshape immune landscapes, and rejuvenate antitumor immunity, offering potential therapeutic avenues in cancer treatment.
Collapse
Affiliation(s)
- Yongmei Wu
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
| | - Wenqian Song
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
| | - Min Su
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
- Center for Tissue Engineering and Stem Cell Research, Key Laboratory of Regenerative Medicine in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| | - Jing He
- Characteristic Key Laboratory of Translational Medicine Research of Cardiovascular and Cerebrovascular Diseases in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| | - Rong Hu
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
- Characteristic Key Laboratory of Translational Medicine Research of Cardiovascular and Cerebrovascular Diseases in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| | - Youbo Zhao
- Department of Human Histology and EmbryologyGuizhou Medical UniversityGuiyangGuizhouChina
- Center for Tissue Engineering and Stem Cell Research, Key Laboratory of Regenerative Medicine in Guizhou ProvinceGuizhou Medical UniversityGuiyangGuizhouChina
| |
Collapse
|
4
|
Zhang J, Zhang Y, Mohibi S, Perng V, Bustamante M, Shi Y, Nakajima K, Chen M, Chen X. Ferredoxin 2 Is Critical for Tumor Suppression and Lipid Homeostasis but Dispensable for Embryonic Development. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:705-716. [PMID: 39732391 DOI: 10.1016/j.ajpath.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/31/2024] [Accepted: 12/03/2024] [Indexed: 12/30/2024]
Abstract
Ferredoxin 1 and 2 (FDX1/2) constitute an evolutionarily conserved FDX family of iron-sulfur cluster-containing proteins. FDX1/2 are cognate substrates of ferredoxin reductase and serve as conduits for electron transfer from NADPH to a set of proteins involved in biogenesis of corticosteroids, hemes, iron-sulfur cluster, and lipoylated proteins. Fdx1 is essential for embryonic development and lipid homeostasis. Herein, Fdx2-deficient mice were generated to explore the physiological role of FDX2. Interestingly, unlike Fdx1-null embryos, which were dead at embryonic day 10.5 to 13.5, Fdx2-null mice were viable. Both Fdx2-null and Fdx2-heterozygous mice had a short lifespan and were susceptible to spontaneous tumors and steatohepatitis. Moreover, FDX2 deficiency increased, whereas overexpression of FDX2 decreased cytoplasmic accumulation of lipid droplets. Consistently, FDX2 deficiency led to accumulation of cholesterol and triglycerides. Mechanistically, FDX2 deficiency suppressed expression of cholesterol transporter ATP-binding cassette transporter A1 (ABCA1) and activated master lipid transcription regulators sterol regulatory element-binding proteins 1/2, thus leading to altered lipid metabolism. Untargeted lipidomic analysis showed that FDX2 deficiency led to altered biosynthesis of various lipid classes, including cardiolipins, cholesterol, ceramides, triglycerides, and fatty acids. In summary, these findings underline an indispensable role of FDX2 in tumor suppression and lipid homeostasis at both cellular and organismal levels without being a prerequisite for embryonic development.
Collapse
Affiliation(s)
- Jin Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, California.
| | - Yanhong Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, California
| | - Shakur Mohibi
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, California
| | - Vivian Perng
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, California
| | - Miranda Bustamante
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, California
| | - Yang Shi
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, California
| | - Kenichi Nakajima
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, California
| | - Mingyi Chen
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Xinbin Chen
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, California.
| |
Collapse
|
5
|
Yang Q, Li Y, Wang X, Ding Q, Tao Y, Li P, Lian X, Chen Y, Zhao L. A high cholesterol diet aggravates experimental colitis through SREBP2-modulated endocytosis and degradation of occludin and Zo-1 proteins. FEBS J 2025; 292:1052-1069. [PMID: 39279038 DOI: 10.1111/febs.17269] [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: 01/11/2024] [Revised: 06/01/2024] [Accepted: 09/02/2024] [Indexed: 09/18/2024]
Abstract
Disrupted cholesterol homeostasis plays a critical role in the development of multiple diseases, such as cardiovascular disease and cancer. However, the role of cholesterol in inflammatory bowel disease (IBD) remains unclear. In the present study, we investigated whether and how high levels of cholesterol in the diet affect experimental colitis in mice. A normal diet supplemented with 1.25% cholesterol (high cholesterol diet) caused more severe colitis and aggravated the disruption of intestinal tight junction structure, accompanied by higher colonic tissue total cholesterol (TC) levels in a dextran sulfate sodium (DSS)-induced experimental colitis mouse model. Cholesterol aggravated DSS-induced intestinal epithelial barrier impairment and nuclear sterol regulatory element-binding protein 2 (nSREBP2) inhibition both in vivo and in vitro. In addition, nSREBP2 overexpression ameliorated cholesterol-induced intestinal epithelial barrier disruption in Caco2 cells. Interestingly, inhibition of SREBP2 disrupted intestinal epithelial barrier in the absence of cholesterol. Furthermore, SREBP2 regulated the protein expression of tight junction proteins (occludin/Zo-1) via modulating caveolin-1-mediated endocytosis and lysosomal degradation. Analysis of UK Biobank data indicated that, in fully adjusted models, higher serum TC concentrations were an independent protective factor for IBD incidence. The sterol regulatory element-binding factor 2 (SREBF2) gene rs2228313 (G/C) genetic variant was associated with the incidence of IBD and the CC genotype of SREBF2 rs2228313 was associated with higher serum TC levels and decreased the risk of IBD. In summary, a high cholesterol diet aggravates DSS-induced colitis in mice by down-regulating nSREBP2 expression, thereby promoting the endocytic degradation of tight junction proteins. In humans, SREBF2 gene single nucleotide polymorphism rs2228313 and serum TC levels are associated with IBD incidence.
Collapse
Affiliation(s)
- Qin Yang
- Centre for Lipid Research & Chongqing Key Laboratory of Metabolism on Lipid and Glucose, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, China
| | - Yongjia Li
- Centre for Lipid Research & Chongqing Key Laboratory of Metabolism on Lipid and Glucose, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, China
| | - Xingxing Wang
- Centre for Lipid Research & Chongqing Key Laboratory of Metabolism on Lipid and Glucose, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, China
| | - Qiuying Ding
- Centre for Lipid Research & Chongqing Key Laboratory of Metabolism on Lipid and Glucose, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, China
| | - Yi Tao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, China
| | - Pan Li
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, China
| | - Xuemei Lian
- Centre for Lipid Research & Chongqing Key Laboratory of Metabolism on Lipid and Glucose, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, China
- School of Public Health, Chongqing Medical University, China
| | - Yaxi Chen
- Centre for Lipid Research & Chongqing Key Laboratory of Metabolism on Lipid and Glucose, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, China
| | - Lei Zhao
- Centre for Lipid Research & Chongqing Key Laboratory of Metabolism on Lipid and Glucose, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, China
| |
Collapse
|
6
|
Yang F, Hu F, Song H, Li T, Xu F, Xu J, Wang L, Wang F, Zhu Y, Huang M, Gao Y, Rao M, Ma H, Tan G. Cholesterol metabolism regulator SREBP2 inhibits HBV replication via suppression of HBx nuclear translocation. Front Immunol 2025; 15:1519639. [PMID: 39872518 PMCID: PMC11769810 DOI: 10.3389/fimmu.2024.1519639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 12/16/2024] [Indexed: 01/30/2025] Open
Abstract
The intricate link between cholesterol metabolism and host immune responses is well recognized, but the specific mechanisms by which cholesterol biosynthesis influences hepatitis B virus (HBV) replication remain unclear. In this study, we show that SREBP2, a key regulator of cholesterol metabolism, inhibits HBV replication by interacting directly with the HBx protein, thereby preventing its nuclear translocation. We also found that inhibiting the ER-to-Golgi transport of the SCAP-SREBP2 complex or blocking SREBP2 maturation significantly enhances HBV suppression. Notably, we demonstrate that the C-terminal domain (CTD) of SREBP2, rather than its N-terminal domain (NTD), mediates this inhibition by interacting with HBx and promoting its extracellular secretion, thus reducing nuclear HBx accumulation. These findings reveal a novel regulatory pathway that links cholesterol metabolism to HBV replication via SREBP2-mediated control of HBx localization. This insight provides a potential basis for new therapeutic strategies against HBV infection, addressing an important global health issue.
Collapse
Affiliation(s)
- Fan Yang
- Department of Hepatology, Center for Pathogen Biology and Infectious Diseases, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
- Department of Anesthesiology, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Feng Hu
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, China
| | - Hongxiao Song
- Department of Hepatology, Center for Pathogen Biology and Infectious Diseases, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Tie Li
- Department of Rheumatology and Immunology, The First Hospital of Jilin University, Changchun, China
| | - Fengchao Xu
- Department of Hepatology, Center for Pathogen Biology and Infectious Diseases, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jing Xu
- Health Examination Center, The First Hospital of Jilin University, Changchun, China
| | - Le Wang
- Department of Hepatology, Center for Pathogen Biology and Infectious Diseases, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Fei Wang
- Department of Pediatrics, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Yujia Zhu
- Department of Hepatology, Center for Pathogen Biology and Infectious Diseases, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Mian Huang
- Department of Hepatology, Center for Pathogen Biology and Infectious Diseases, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yanli Gao
- Department of Pediatrics, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Min Rao
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, China
| | - Haichun Ma
- Department of Anesthesiology, The First Hospital, Jilin University, Changchun, Jilin, China
| | - Guangyun Tan
- Department of Hepatology, Center for Pathogen Biology and Infectious Diseases, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
7
|
Taniguchi M. Utilizing Indigenous Animal Genetic Resources-Based on Research Into Indigenous Cattle Breeds in the Basque Country in Northern Spain and Indigenous Pig Breeds in Vietnam. Anim Sci J 2025; 96:e70046. [PMID: 40069921 PMCID: PMC11897423 DOI: 10.1111/asj.70046] [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: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 03/15/2025]
Abstract
Biodiversity, climate change, and food security are closely related and increasing worldwide concerns. Therefore, sustainable productivity and changes to the livestock industry are required for the maintenance or amelioration of the global environment and the future of humanity. This review describes the potential of animal genetic resources and their expected roles in livestock improvement. Herein, I report the findings of my previous collaborative research project on the domestication of cattle and pigs, and genetic analysis of native cattle in the Basque Country, Spain, to improve the meat quality of native livestock genetic resources. In addition, I review another research on the diversity of native pigs in Vietnam and the establishment of a gene bank. The conservation of native livestock genetic resources is important for sustainable food production in each region, the inheritance of food culture, and to be available for future needs because native livestock adapted to their habitat's environment can coexist with locally cultivated crops. This encourages livestock researchers to consider sustainable production through the future use of native livestock genetic resources and to seek feasible solutions.
Collapse
Affiliation(s)
- Masaaki Taniguchi
- Institute of Agrobiological SciencesNational Agriculture and Food Research OrganizationTsukubaJapan
| |
Collapse
|
8
|
Cheng Y, Jung J, Guo L, Shuboni-Mulligan DD, Chen JF, Hu W, Guo ML. HIV-TAT dysregulates microglial lipid metabolism through SREBP2/miR-124 axis: Implication of lipid droplet accumulation microglia in NeuroHIV. Brain Behav Immun 2025; 123:108-122. [PMID: 39260763 PMCID: PMC11624073 DOI: 10.1016/j.bbi.2024.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 08/26/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024] Open
Abstract
Chronic HIV infection can dysregulate lipid/cholesterol metabolism in the peripheral system, contributing to the higher incidences of diabetes and atherosclerosis in HIV (+) individuals. Recently, accumulating evidence indicate that HIV proteins can also dysregulate lipid/cholesterol metabolism in the brain and such dysregulation could be linked with the pathogenesis of HIV-associated neurological disorders (HAND)/NeuroHIV. To further characterize the association between lipid/cholesterol metabolism and HAND, we employed HIV-inducible transactivator of transcription (iTAT) and control mice to compare their brain lipid profiles. Our results reveal that HIV-iTAT mice possess dysregulated lipid profiles and have increased numbers of lipid droplets (LDs) accumulation microglia (LDAM) in the brains. HIV protein TAT can upregulate LDs formation through enhancing the lipid/cholesterol synthesis in vitro. Mechanistically, HIV-TAT increases the expression of sterol regulatory element-binding protein 2 (SREBP2) through microRNA-124 downregulation. Cholesterol synthesis inhibition can block HIV-TAT-mediated NLRP3 inflammasome activation and microglial activation in vitro as well as mitigate aging-related behavioral impairment and memory deficiency in HIV-iTAT mice. Taken together, our results indicate an inherent role of lipid metabolism and LDAM in the pathogenesis of NeuroHIV (immunometabolism). These findings suggest that LDAM reversal through modulating lipid/cholesterol metabolism could be a novel therapeutic target for ameliorating NeuroHIV symptoms in chronic HIV (+) individuals.
Collapse
Affiliation(s)
- Yan Cheng
- Department of Biomedical and Translational Sciences, Macro & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - Jaekeun Jung
- Department of Biomedical and Translational Sciences, Macro & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - Liyang Guo
- Department of Biomedical and Translational Sciences, Macro & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - Dorela D Shuboni-Mulligan
- Department of Biomedical and Translational Sciences, Macro & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA
| | - Jian-Fu Chen
- Center for Craniofacial Molecular Biology, University of Southern California (USC), Los Angeles, CA 90033, USA
| | - Wenhui Hu
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Ming-Lei Guo
- Department of Biomedical and Translational Sciences, Macro & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA 23507, USA; Center for Integrative Neuroscience and Inflammatory Diseases, Macro & Joan Brock Virginia Health Science, Old Dominion University, Norfolk, VA 23507, USA.
| |
Collapse
|
9
|
Ling L, Li R, Xu M, Zhou J, Hu M, Zhang X, Zhang XJ. Species differences of fatty liver diseases: comparisons between human and feline. Am J Physiol Endocrinol Metab 2025; 328:E46-E61. [PMID: 39636211 DOI: 10.1152/ajpendo.00014.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as the most widespread chronic liver disease that poses significant threats to public health due to changes in dietary habits and lifestyle patterns. The transition from simple steatosis to nonalcoholic steatohepatitis (NASH) markedly increases the risk of developing cirrhosis, hepatocellular carcinoma, and liver failure in patients. However, there is only one Food and Drug Administration-approved therapeutic drug in the world, and the clinical demand is huge. There is significant clinical heterogeneity among patients with NAFLD, and it is challenging to fully understand human NAFLD using only a single animal model. Interestingly, felines, like humans, are particularly prone to spontaneous fatty liver disease. This review summarized and compared the etiology, clinical features, pathological characteristics, and molecular pathogenesis between human fatty liver and feline hepatic lipidosis (FHL). We analyzed the key similarities and differences between those two species, aiming to provide theoretical foundations for developing effective strategies for the treatment of NAFLD in clinics.
Collapse
Affiliation(s)
- Like Ling
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Ruilin Li
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Mengqiong Xu
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Junjie Zhou
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Manli Hu
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Xin Zhang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Xiao-Jing Zhang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Piombo E, Tzelepis G, Ruus AG, Rafiei V, Jensen DF, Karlsson M, Dubey M. Sterol regulatory element-binding proteins mediate intrinsic fungicide tolerance and antagonism in the fungal biocontrol agent Clonostachys rosea IK726. Microbiol Res 2024; 289:127922. [PMID: 39368255 DOI: 10.1016/j.micres.2024.127922] [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: 07/01/2024] [Revised: 09/10/2024] [Accepted: 09/25/2024] [Indexed: 10/07/2024]
Abstract
Sterol regulatory element-binding proteins (SREBPs) are transcription factors governing various biological processes in fungi, including virulence and fungicide tolerance, by regulating ergosterol biosynthesis and homeostasis. While studied in model fungal species, their role in fungal species used for biocontrol remains elusive. This study delves into the biological and regulatory function of SREBPs in the fungal biocontrol agent (BCA) Clonostachys rosea IK726, with a specific focus on fungicide tolerance and antagonism. Clonostachys rosea genome contains two SREBP coding genes (sre1 and sre2) with distinct characteristics. Deletion of sre1 resulted in mutant strains with pleiotropic phenotypes, including reduced C. rosea growth on medium supplemented with prothioconazole and boscalid fungicides, hypoxia mimicking agent CoCl2 and cell wall stressor SDS, and altered antagonistic abilities against Botrytis cinerea and Rhizoctonia solani. However, Δsre2 strains showed no significant effect. Consistent with the gene deletion results, overexpression of sre1 in Saccharomyces cerevisiae enhanced tolerance to prothioconazole. The functional differentiation between SRE1 and SRE2 was elucidated by the yeast-two-hybridization assay, which showed an interaction between SREBP cleavage-activating protein (SCAP) and SRE1 but not between SRE2 and SCAP. Transcriptome analysis of the Δsre1 strain unveiled SRE1-mediated expression regulation of genes involved in lipid metabolism, respiration, and xenobiotic tolerance. Notably, genes coding for antimicrobial compounds chitinases and polyketide synthases were downregulated, aligning with the altered antagonism phenotype. This study uncovers the role of SREBPs in fungal BCAs, providing insights for C. rosea IK726 application into integrated pest management strategies.
Collapse
Affiliation(s)
- Edoardo Piombo
- Department of Forest Mycology and Plant Pathology, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Georgios Tzelepis
- Department of Forest Mycology and Plant Pathology, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Alma Gustavsson Ruus
- Department of Forest Mycology and Plant Pathology, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Vahideh Rafiei
- Department of Forest Mycology and Plant Pathology, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Dan Funck Jensen
- Department of Forest Mycology and Plant Pathology, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Magnus Karlsson
- Department of Forest Mycology and Plant Pathology, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Mukesh Dubey
- Department of Forest Mycology and Plant Pathology, Swedish University of Agricultural Sciences, Uppsala, Sweden.
| |
Collapse
|
11
|
Soares VC, Dias SSG, Santos JC, Bozza PT. Unlocking secrets: lipid metabolism and lipid droplet crucial roles in SARS-CoV-2 infection and the immune response. J Leukoc Biol 2024; 116:1254-1268. [PMID: 39087951 DOI: 10.1093/jleuko/qiae170] [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: 01/31/2024] [Revised: 06/11/2024] [Accepted: 07/31/2024] [Indexed: 08/02/2024] Open
Abstract
Lipid droplets (LDs) are crucial for maintaining lipid and energy homeostasis within cells. LDs are highly dynamic organelles that present a phospholipid monolayer rich in neutral lipids. Additionally, LDs are associated with structural and nonstructural proteins, rapidly mobilizing lipids for various biological processes. Lipids play a pivotal role during viral infection, participating during viral membrane fusion, viral replication, and assembly, endocytosis, and exocytosis. SARS-CoV-2 infection often induces LD accumulation, which is used as a source of energy for the replicative process. These findings suggest that LDs are a hallmark of viral infection, including SARS-CoV-2 infection. Moreover, LDs participate in the inflammatory process and cell signaling, activating pathways related to innate immunity and cell death. Accumulating evidence demonstrates that LD induction by SARS-CoV-2 is a highly coordinated process, aiding replication and evading the immune system, and may contribute to the different cell death process observed in various studies. Nevertheless, recent research in the field of LDs suggests these organelles according to the pathogen and infection conditions may also play roles in immune and inflammatory responses, protecting the host against viral infection. Understanding how SARS-CoV-2 influences LD biogenesis is crucial for developing novel drugs or repurposing existing ones. By targeting host lipid metabolic pathways exploited by the virus, it is possible to impact viral replication and inflammatory responses. This review seeks to discuss and analyze the role of LDs during SARS-CoV-2 infection, specifically emphasizing their involvement in viral replication and the inflammatory response.
Collapse
Affiliation(s)
- Vinicius Cardoso Soares
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Program of Immunology and Inflammation, Federal University of Rio de Janeiro, UFRJ, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Suelen Silva Gomes Dias
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Julia Cunha Santos
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| | - Patrícia T Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
- Center for Research, Innovation and Surveillance in COVID-19 and Heath Emergencies, Oswaldo Cruz Foundation, Fiocruz, Brasil Ave, Rio de Janeiro, RJ, 21040-361, Brazil
| |
Collapse
|
12
|
Durán A, Venegas M, Barahona S, Sepúlveda D, Baeza M, Cifuentes V, Alcaíno J. Increasing carotenoid production in Xanthophyllomyces dendrorhous/Phaffia rhodozyma: SREBP pathway activation and promoter engineering. Biol Res 2024; 57:78. [PMID: 39497228 PMCID: PMC11536662 DOI: 10.1186/s40659-024-00559-1] [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: 01/22/2024] [Accepted: 10/24/2024] [Indexed: 11/07/2024] Open
Abstract
The yeast Xanthophyllomyces dendrorhous synthesizes astaxanthin, a high-value carotenoid with biotechnological relevance in the nutraceutical and aquaculture industries. However, enhancing carotenoid production through strain engineering remains an ongoing challenge. Recent studies have demonstrated that carotenogenesis in X. dendrorhous is regulated by the SREBP pathway, which includes the transcription factor Sre1, particularly in the mevalonate pathway that also produces precursors used for ergosterol synthesis. In this study, we explored a novel approach to enhance carotenoid synthesis by replacing the native crtE promoter, which drives geranylgeranyl pyrophosphate synthesis (the step where carotenogenesis diverges from ergosterol biosynthesis), with the promoter of the HMGS gene, which encodes 3-hydroxy-3-methylglutaryl-CoA synthase from the mevalonate pathway. The impact of this substitution was evaluated in two mutant strains that already overproduce carotenoids due to the presence of an active Sre1 transcription factor: CBS.cyp61-, which does not produce ergosterol and strain CBS.SRE1N.FLAG, which constitutively expresses the active form of Sre1. Wild-type strain CBS6938 was used as a control. Our results showed that this modification increased the crtE transcript levels more than threefold and fourfold in CBS.cyp61-.pHMGS/crtE and CBS.SRE1N.FLAG.pHMGS/crtE, respectively, resulting in 1.43-fold and 1.22-fold increases in carotenoid production. In contrast, this modification did not produce significant changes in the wild-type strain, which lacks the active Sre1 transcription factor under the same culture conditions. This study highlights the potential of promoter substitution strategies involving genes regulated by Sre1 to enhance carotenoid production, specifically in strains where the SREBP pathway is activated, offering a promising avenue for strain improvement in industrial applications.
Collapse
Affiliation(s)
- Alejandro Durán
- Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Santiago, Chile
| | - Maximiliano Venegas
- Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Santiago, Chile
| | - Salvador Barahona
- Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Santiago, Chile
| | - Dionisia Sepúlveda
- Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Santiago, Chile
| | - Marcelo Baeza
- Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Santiago, Chile
| | - Víctor Cifuentes
- Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Santiago, Chile
| | - Jennifer Alcaíno
- Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Las Palmeras 3425, Santiago, Chile.
| |
Collapse
|
13
|
Garmo LC, Herroon MK, Mecca S, Wilson A, Allen DR, Agarwal M, Kim S, Petriello MC, Podgorski I. The long-chain polyfluorinated alkyl substance perfluorohexane sulfonate (PFHxS) promotes bone marrow adipogenesis. Toxicol Appl Pharmacol 2024; 491:117047. [PMID: 39111555 DOI: 10.1016/j.taap.2024.117047] [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: 05/17/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/11/2024]
Abstract
Per- and polyfluoroalkyl substances (PFAS) bioaccumulate in different organ systems, including bone. While existing research highlights the adverse impact of PFAS on bone density, a critical gap remains in understanding the specific effects on the bone marrow microenvironment, especially the bone marrow adipose tissue (BMAT). Changes in BMAT have been linked to various health consequences, such as the development of osteoporosis and the progression of metastatic tumors in bone. Studies presented herein demonstrate that exposure to a mixture of five environmentally relevant PFAS compounds promotes marrow adipogenesis in vitro and in vivo. We show that among the components of the mixture, PFHxS, an alternative to PFOS, has the highest propensity to accumulate in bone and effectively promote marrow adipogenesis. Utilizing RNAseq approaches, we identified the peroxisome proliferator-activated receptor (PPAR) signaling as a top pathway modulated by PFHxS exposure. Furthermore, we provide results suggesting the activation and involvement of PPAR-gamma (PPARγ) in PFHxS-mediated bone marrow adipogenesis, especially in combination with high-fat diet. In conclusion, our findings demonstrate the potential impact of elevated PFHxS levels, particularly in occupational settings, on bone health, and specifically bone marrow adiposity. This study contributes new insights into the health risks of PFHxS exposure, urging further research on the relationship between environmental factors, diet, and adipose tissue dynamics.
Collapse
Affiliation(s)
- Laimar C Garmo
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Mackenzie K Herroon
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Shane Mecca
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Alexis Wilson
- Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, United States of America
| | - David R Allen
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Manisha Agarwal
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Seongho Kim
- Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, United States of America
| | - Michael C Petriello
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America; Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, United States of America
| | - Izabela Podgorski
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America; Department of Oncology, Wayne State University School of Medicine and Karmanos Cancer Institute, Detroit, MI, United States of America.
| |
Collapse
|
14
|
Moulton MJ, Atala K, Zheng Y, Dutta D, Grange DK, Lin WW, Wegner DJ, Wambach JA, Duker AL, Bober MB, Kratz L, Wise CA, Oxendine I, Khanshour A, Wangler MF, Yamamoto S, Cole FS, Rios J, Bellen HJ. Dominant missense variants in SREBF2 are associated with complex dermatological, neurological, and skeletal abnormalities. Genet Med 2024; 26:101174. [PMID: 38847193 DOI: 10.1016/j.gim.2024.101174] [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: 01/30/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 07/21/2024] Open
Abstract
PURPOSE We identified 2 individuals with de novo variants in SREBF2 that disrupt a conserved site 1 protease (S1P) cleavage motif required for processing SREBP2 into its mature transcription factor. These individuals exhibit complex phenotypic manifestations that partially overlap with sterol regulatory element binding proteins (SREBP) pathway-related disease phenotypes, but SREBF2-related disease has not been previously reported. Thus, we set out to assess the effects of SREBF2 variants on SREBP pathway activation. METHODS We undertook ultrastructure and gene expression analyses using fibroblasts from an affected individual and utilized a fly model of lipid droplet (LD) formation to investigate the consequences of SREBF2 variants on SREBP pathway function. RESULTS We observed reduced LD formation, endoplasmic reticulum expansion, accumulation of aberrant lysosomes, and deficits in SREBP2 target gene expression in fibroblasts from an affected individual, indicating that the SREBF2 variant inhibits SREBP pathway activation. Using our fly model, we discovered that SREBF2 variants fail to induce LD production and act in a dominant-negative manner, which can be rescued by overexpression of S1P. CONCLUSION Taken together, these data reveal a mechanism by which SREBF2 pathogenic variants that disrupt the S1P cleavage motif cause disease via dominant-negative antagonism of S1P, limiting the cleavage of S1P targets, including SREBP1 and SREBP2.
Collapse
Affiliation(s)
- Matthew J Moulton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX
| | - Kristhen Atala
- Center for Translational Research, Scottish Rite for Children, Dallas, TX
| | - Yiming Zheng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX; Current address: State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Debdeep Dutta
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX
| | - Dorothy K Grange
- Edward Mallinckrodt Department of Pediatrics, Washington University in St. Louis School of Medicine and St. Louis Children's Hospital, St. Louis, MO
| | - Wen-Wen Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX
| | - Daniel J Wegner
- Edward Mallinckrodt Department of Pediatrics, Washington University in St. Louis School of Medicine and St. Louis Children's Hospital, St. Louis, MO
| | - Jennifer A Wambach
- Edward Mallinckrodt Department of Pediatrics, Washington University in St. Louis School of Medicine and St. Louis Children's Hospital, St. Louis, MO
| | - Angela L Duker
- Skeletal Dysplasia Program, Orthogenetics, Nemours Children's Hospital, Wilmington, DE
| | - Michael B Bober
- Skeletal Dysplasia Program, Orthogenetics, Nemours Children's Hospital, Wilmington, DE
| | - Lisa Kratz
- Kennedy Krieger Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Carol A Wise
- Center for Translational Research, Scottish Rite for Children, Dallas, TX; Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX; Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX; Department of Orthopedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - Ila Oxendine
- Center for Translational Research, Scottish Rite for Children, Dallas, TX
| | - Anas Khanshour
- Center for Translational Research, Scottish Rite for Children, Dallas, TX
| | - Michael F Wangler
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX
| | - F Sessions Cole
- Edward Mallinckrodt Department of Pediatrics, Washington University in St. Louis School of Medicine and St. Louis Children's Hospital, St. Louis, MO
| | - Jonathan Rios
- Center for Translational Research, Scottish Rite for Children, Dallas, TX; Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX; Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX; Department of Orthopedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX.
| |
Collapse
|
15
|
Chacón CF, Parachú Marcó MV, Poletta GL, Siroski PA. Lipid metabolism in crocodilians: A field with promising applications in the field of ecotoxicology. ENVIRONMENTAL RESEARCH 2024; 252:119017. [PMID: 38704009 DOI: 10.1016/j.envres.2024.119017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024]
Abstract
In the last years, lipid physiology has become an important research target for systems biology applied to the field of ecotoxicology. Lipids are not only essential components of biological membranes, but also participate in extra and intracellular signaling processes and as signal transducers and amplifiers of regulatory cascades. Particularly in sauropsids, lipids are the main source of energy for reproduction, growth, and embryonic development. In nature, organisms are exposed to different stressors, such as parasites, diseases and environmental contaminants, which interact with lipid signaling and metabolic pathways, disrupting lipid homeostasis. The system biology approach applied to ecotoxicological studies is crucial to evaluate metabolic regulation under environmental stress produced by xenobiotics. In this review, we cover information of molecular mechanisms that contribute to lipid metabolism homeostasis in sauropsids, specifically in crocodilian species. We focus on the role of lipid metabolism as a powerful source of energy and its importance during oocyte maturation, which has been increasingly recognized in many species, but information is still scarce in crocodiles. Finally, we highlight priorities for future research on the influence of environmental stressors on lipid metabolism, their potential effect on the reproductive system and thus on the offspring, and their implications on crocodilians conservation.
Collapse
Affiliation(s)
- C F Chacón
- Laboratorio de Ecología Molecular Aplicada (LEMA), Instituto de Ciencias Veterinarias del Litoral- Consejo Nacional de Investigaciones Científicas y Técnicas (ICiVet Litoral-CONICET/UNL), Av. Aristóbulo del Valle 8700, 3000, Santa Fe, Argentina; Proyecto Yacaré (MAyCC, Gob. de Santa Fe), Av. Aristóbulo del Valle 8700, 3000, Santa Fe, Argentina.
| | - M V Parachú Marcó
- Laboratorio de Ecología Molecular Aplicada (LEMA), Instituto de Ciencias Veterinarias del Litoral- Consejo Nacional de Investigaciones Científicas y Técnicas (ICiVet Litoral-CONICET/UNL), Av. Aristóbulo del Valle 8700, 3000, Santa Fe, Argentina; Proyecto Yacaré (MAyCC, Gob. de Santa Fe), Av. Aristóbulo del Valle 8700, 3000, Santa Fe, Argentina
| | - G L Poletta
- Laboratorio de Ecología Molecular Aplicada (LEMA), Instituto de Ciencias Veterinarias del Litoral- Consejo Nacional de Investigaciones Científicas y Técnicas (ICiVet Litoral-CONICET/UNL), Av. Aristóbulo del Valle 8700, 3000, Santa Fe, Argentina; Toxicología, Farmacología y Bioquímica Legal, FBCB-UNL, CONICET, Ciudad Universitaria, Paraje El Pozo S/N, 3000, Santa Fe, Argentina
| | - P A Siroski
- Laboratorio de Ecología Molecular Aplicada (LEMA), Instituto de Ciencias Veterinarias del Litoral- Consejo Nacional de Investigaciones Científicas y Técnicas (ICiVet Litoral-CONICET/UNL), Av. Aristóbulo del Valle 8700, 3000, Santa Fe, Argentina; Proyecto Yacaré (MAyCC, Gob. de Santa Fe), Av. Aristóbulo del Valle 8700, 3000, Santa Fe, Argentina
| |
Collapse
|
16
|
Zhu Y, Cho K, Lacin H, Zhu Y, DiPaola JT, Wilson BA, Patti GJ, Skeath JB. Loss of dihydroceramide desaturase drives neurodegeneration by disrupting endoplasmic reticulum and lipid droplet homeostasis in glial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.01.573836. [PMID: 38260379 PMCID: PMC10802327 DOI: 10.1101/2024.01.01.573836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Dihydroceramide desaturases convert dihydroceramides to ceramides, the precursors of all complex sphingolipids. Reduction of DEGS1 dihydroceramide desaturase function causes pediatric neurodegenerative disorder hypomyelinating leukodystrophy-18 (HLD-18). We discovered that infertile crescent (ifc), the Drosophila DEGS1 homolog, is expressed primarily in glial cells to promote CNS development by guarding against neurodegeneration. Loss of ifc causes massive dihydroceramide accumulation and severe morphological defects in cortex glia, including endoplasmic reticulum (ER) expansion, failure of neuronal ensheathment, and lipid droplet depletion. RNAi knockdown of the upstream ceramide synthase schlank in glia of ifc mutants rescues ER expansion, suggesting dihydroceramide accumulation in the ER drives this phenotype. RNAi knockdown of ifc in glia but not neurons drives neuronal cell death, suggesting that ifc function in glia promotes neuronal survival. Our work identifies glia as the primary site of disease progression in HLD-18 and may inform on juvenile forms of ALS, which also feature elevated dihydroceramide levels.
Collapse
Affiliation(s)
- Yuqing Zhu
- Department of Genetics, Washington University School of Medicine, 4523 Clayton Avenue, St. Louis, MO 63110, USA
| | - Kevin Cho
- Department of Chemistry, Washington University in St. Louis, One Brookings Drive, St. Louis, MO 63130, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University in St. Louis, One Brookings Drive, St. Louis, MO 63130, USA
| | - Haluk Lacin
- Division of Biological and Biomedical Systems, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Yi Zhu
- Department of Genetics, Washington University School of Medicine, 4523 Clayton Avenue, St. Louis, MO 63110, USA
| | - Jose T DiPaola
- Department of Genetics, Washington University School of Medicine, 4523 Clayton Avenue, St. Louis, MO 63110, USA
| | - Beth A Wilson
- Department of Genetics, Washington University School of Medicine, 4523 Clayton Avenue, St. Louis, MO 63110, USA
| | - Gary J Patti
- Department of Chemistry, Washington University in St. Louis, One Brookings Drive, St. Louis, MO 63130, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Mass Spectrometry and Metabolic Tracing, Washington University in St. Louis, One Brookings Drive, St. Louis, MO 63130, USA
| | - James B Skeath
- Department of Genetics, Washington University School of Medicine, 4523 Clayton Avenue, St. Louis, MO 63110, USA
| |
Collapse
|
17
|
Ma S, Ming Y, Wu J, Cui G. Cellular metabolism regulates the differentiation and function of T-cell subsets. Cell Mol Immunol 2024; 21:419-435. [PMID: 38565887 PMCID: PMC11061161 DOI: 10.1038/s41423-024-01148-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 02/23/2024] [Indexed: 04/04/2024] Open
Abstract
T cells are an important component of adaptive immunity and protect the host from infectious diseases and cancers. However, uncontrolled T cell immunity may cause autoimmune disorders. In both situations, antigen-specific T cells undergo clonal expansion upon the engagement and activation of antigens. Cellular metabolism is reprogrammed to meet the increase in bioenergetic and biosynthetic demands associated with effector T cell expansion. Metabolites not only serve as building blocks or energy sources to fuel cell growth and expansion but also regulate a broad spectrum of cellular signals that instruct the differentiation of multiple T cell subsets. The realm of immunometabolism research is undergoing swift advancements. Encapsulating all the recent progress within this concise review in not possible. Instead, our objective is to provide a succinct introduction to this swiftly progressing research, concentrating on the metabolic intricacies of three pivotal nutrient classes-lipids, glucose, and amino acids-in T cells. We shed light on recent investigations elucidating the roles of these three groups of metabolites in mediating the metabolic and immune functions of T cells. Moreover, we delve into the prospect of "editing" metabolic pathways within T cells using pharmacological or genetic approaches, with the aim of synergizing this approach with existing immunotherapies and enhancing the efficacy of antitumor and antiinfection immune responses.
Collapse
Affiliation(s)
- Sicong Ma
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China
| | - Yanan Ming
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China
| | - Jingxia Wu
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China.
| | - Guoliang Cui
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230601, China.
| |
Collapse
|
18
|
Zhu W, Hong Y, Tong Z, He X, Li Y, Wang H, Gao X, Song P, Zhang X, Wu X, Tan Z, Huang W, Liu Z, Bao Y, Ma J, Zheng N, Xie C, Ke X, Zhou W, Jia W, Li M, Zhong J, Sheng L, Li H. Activation of hepatic adenosine A1 receptor ameliorates MASH via inhibiting SREBPs maturation. Cell Rep Med 2024; 5:101477. [PMID: 38508143 PMCID: PMC10983109 DOI: 10.1016/j.xcrm.2024.101477] [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: 06/26/2023] [Revised: 12/10/2023] [Accepted: 02/21/2024] [Indexed: 03/22/2024]
Abstract
Metabolic (dysfunction)-associated steatohepatitis (MASH) is the advanced stage of metabolic (dysfunction)-associated fatty liver disease (MAFLD) lacking approved clinical drugs. Adenosine A1 receptor (A1R), belonging to the G-protein-coupled receptors (GPCRs) superfamily, is mainly distributed in the central nervous system and major peripheral organs with wide-ranging physiological functions; however, the exact role of hepatic A1R in MAFLD remains unclear. Here, we report that liver-specific depletion of A1R aggravates while overexpression attenuates diet-induced metabolic-associated fatty liver (MAFL)/MASH in mice. Mechanistically, activation of hepatic A1R promotes the competitive binding of sterol-regulatory element binding protein (SREBP) cleavage-activating protein (SCAP) to sequestosome 1 (SQSTM1), rather than protein kinase A (PKA) leading to SCAP degradation in lysosomes. Reduced SCAP hinders SREBP1c/2 maturation and thus suppresses de novo lipogenesis and inflammation. Higher hepatic A1R expression is observed in patients with MAFL/MASH and high-fat diet (HFD)-fed mice, which is supposed to be a physiologically adaptive response because A1R agonists attenuate MAFL/MASH in an A1R-dependent manner. These results highlight that hepatic A1R is a potential target for MAFL/MASH therapy.
Collapse
Affiliation(s)
- Weize Zhu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ying Hong
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhaowei Tong
- Huzhou Key Laboratory of Precision Medicine Research and Translation for Infectious Diseases, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou 313000, China
| | - Xiaofang He
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yan Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hao Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xinxin Gao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Pengtao Song
- Department of Pathology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou 313000, China
| | - Xianshan Zhang
- Huzhou Key Laboratory of Precision Medicine Research and Translation for Infectious Diseases, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou 313000, China
| | - Xiaochang Wu
- Department of Hepatobiliary Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou 313000, China
| | - Zhenhua Tan
- Department of Hepatobiliary Surgery, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou 313000, China
| | - Wenjin Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zekun Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yiyang Bao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Junli Ma
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ningning Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Cen Xie
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xisong Ke
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wen Zhou
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural, Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Wei Jia
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong 999077, China
| | - Mingxiao Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jing Zhong
- Huzhou Key Laboratory of Precision Medicine Research and Translation for Infectious Diseases, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou 313000, China.
| | - Lili Sheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Houkai Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
19
|
Guo ML, Cheng Y, Pineda DM, Dempsey RE, Yang L. Lipid Droplets Accumulation in the Brain of HIV Transgenic Rat: Implication in the Accelerated Aging of HIV Infected Individuals. Aging Dis 2024; 16:AD.2024.0125. [PMID: 38377024 PMCID: PMC11745453 DOI: 10.14336/ad.2024.0125] [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: 12/05/2023] [Accepted: 01/25/2024] [Indexed: 02/22/2024] Open
Abstract
Abnormal microglial activation has been suggested as "driven force" promoting brain aging. Lipid droplets accumulating microglia (LDAM), identified as a novel inflammatory phenotype, elevate neuroinflammation and exaggerate neuronal injuries in aging and multiple neurodegenerative diseases. Since chronic HIV (human immunodeficiency virus) (+) individuals show an accelerated brain aging and higher incidence of neurological symptoms compared to age-matched HIV (-) population, we hypothesize that LDAM are also involved in such phenomenon. For validating the hypothesis, we employed HIV transgenic (HIV-Tg) and wilt type (WT) rats to check lipid droplets (LDs) accumulation in the brains at mature (6 months) and middle age (12 months). Our results showed that HIV-Tg rats possess higher levels of LDs formation in the hippocampus (HP) and prefrontal cortex (PFc) than controls at middle age. Increased LDs are mainly presented in microglia in the HP but largely co-localized with astrocytes in the PFc. Interestingly, increased LDs are associated with upregulation on Iba1 but not with GFAP levels. HIV-Tg rats reveal an accelerated LDs accumulation during normal aging. Purified microglia from HIV-Tg rats (12 month) show higher expression of neuroimmune signaling than microglia from controls. HIV-Tg rats showed dysregulation on cholesterol synthesis in the brain HP as well as deficiency on locomotion coordination compared to controls. Overall, our results demonstrate substantial LDs accumulation in the brains of HIV-Tg rats which is associated with abnormal microglial activation and accelerated decline on locomotion coordination during aging. Dysregulation on lipid metabolism might underlie accelerated brain aging in the context of chronic HIV infection.
Collapse
Affiliation(s)
- Ming-Lei Guo
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, Virginia, USA.
- Center for Integrative Neuroscience and Inflammatory Diseases, Eastern Virginia Medical School, Norfolk, Virginia, USA.
| | - Yan Cheng
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, Virginia, USA.
| | - Damian Martinez Pineda
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, Virginia, USA.
| | - Rachael E. Dempsey
- Drug Addiction Laboratory, Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, Virginia, USA.
| | - Lifang Yang
- Leroy T. Canoles Jr. Cancer Research Center, Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia, USA.
| |
Collapse
|
20
|
Zhou ZQ, Wei M, Tan CL, Deng ZY, Li J. Low intake of ruminant trans fatty acids ameliorates the disordered lipid metabolism in C57BL/6J mice fed a high-fat diet. Food Funct 2024; 15:1539-1552. [PMID: 38234289 DOI: 10.1039/d3fo04947g] [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: 01/19/2024]
Abstract
Currently, the health benefits of ruminant trans fatty acids (R-TFA) are still controversial. Our previous investigations indicated that R-TFA at higher dosages (1.3% and 4% E) caused disordered lipid metabolism in mice; however, through collecting R-TFA intake data in 9 provinces of China, it was suggested that, in 2021, the range of R-TFA intake for Chinese residents was about 0.053-0.307 g d-1. Based on the 2022 Nutritional Dietary Guidelines for Chinese Residents, the recommended daily energy supply from R-TFA was about 0.11%-0.15% E. However, the health effects of R-TFA at a lower dosage are still unknown; therefore, our current research aims to further explore the effects of R-TFA on health. Through in vivo experiments, it was shown that R-TFA (0.15% E) decreased body weight gain and serum cholesterol levels in C57BL/6J mice fed a high-fat diet, while it had no significant effect on mice fed a low-fat diet. Besides, hepatic histopathology analysis suggested that R-TFA (0.15% E) ameliorated the degree of hepatic steatosis and reduced intrahepatocyte lipid droplet accumulation in C57BL/6J mice fed a high-fat diet. Through lipidomics analysis, we further screened 8 potential lipid metabolites that participate in regulating the dysregulation of lipid metabolism. Finally, it was suggested that R-TFA (0.15% E) down-regulated the expression of genes related to inflammation and cholesterol synthesis while up-regulated the expression of genes related to cholesterol clearance, which might partially explain the salutary effect of R-TFA (0.15% E) in ameliorating the hepatic steatosis and improving disordered lipid metabolism in mice fed a high-fat diet. Our current research will provide a reference for the intake of R-TFA and, furthermore, give some insights into understanding the health effects of R-TFA.
Collapse
Affiliation(s)
- Ze-Qiang Zhou
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China.
| | - Meng Wei
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China.
| | - Chao-Li Tan
- Jiangxi Sunshine Dairy Co., Ltd, Nanchang, Jiangxi 330001, China
| | - Ze-Yuan Deng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China.
- College of Food, Nanchang University, Nanchang 330047, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330031, China
| | - Jing Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China.
- College of Food, Nanchang University, Nanchang 330047, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330031, China
- National Center of Technology Innovation for Dairy, China
| |
Collapse
|
21
|
Mendoza-Grimau V, Pérez-Gálvez A, Busturia A, Fontecha J. Lipidomic profiling of Drosophila strains Canton-S and white 1118 reveals intraspecific lipid variations in basal metabolic rate. Prostaglandins Leukot Essent Fatty Acids 2024; 201:102618. [PMID: 38795635 DOI: 10.1016/j.plefa.2024.102618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/23/2024] [Accepted: 05/08/2024] [Indexed: 05/28/2024]
Abstract
Drosophila melanogaster is a well-established model system for studies on lipid metabolism and energy homeostasis. In this study, we identified and quantified the main components of the lipid profile of two widely utilized Drosophila strains, namely Canton-S and white1118, under identical experimental conditions. Differences observed between the strains can be attributed to inherent metabolic divergences, thus limiting the influence of confounding factors. Using the comprehensive lipid data acquired, we applied cluster analysis and PLS-DA techniques to ascertain whether the lipidome could effectively differentiate between the strains. Certain lipid features, such as triacylglycerols, polar lipids, and specific sterol components, could be distinguished between flies of both strains regardless of sex. Our results suggest that although Canton-S and white1118 have similar lipid profiles and distributions, a selected subset of lipids demonstrates clear discriminatory potential between strains, thereby bearing significant implications for planning biological studies using these strains as control references.
Collapse
Affiliation(s)
- Victor Mendoza-Grimau
- Food Lipid Biomarkers and Health Group, Institute of Food Science Research (CIAL, CSIC-UAM), Madrid 28049, Spain
| | - Antonio Pérez-Gálvez
- Group of Chemistry and Biochemistry of Pigments, Instituto de la Grasa, CSIC, Sevilla 41013, Spain
| | - Ana Busturia
- Tissue and organ homeostasis, Centro de Biología Molecular Severo Ochoa, (CBMSO, CSIC-UAM), Madrid 28049, Spain
| | - Javier Fontecha
- Food Lipid Biomarkers and Health Group, Institute of Food Science Research (CIAL, CSIC-UAM), Madrid 28049, Spain.
| |
Collapse
|
22
|
Li J, Wu YJ. Tri-ortho-cresyl phosphate induces hepatic steatosis by mTOR activation and ER stress induction. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 271:116010. [PMID: 38280340 DOI: 10.1016/j.ecoenv.2024.116010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 12/17/2023] [Accepted: 01/21/2024] [Indexed: 01/29/2024]
Abstract
Tri-ortho-cresyl phosphate (TOCP), an organophosphorus compound (OP), which is widely used as plasticizer, flame retardant and other industrial products, has been reported to cause multiple toxicities including neurotoxicity and reproductive toxicity. However, it remains to be elusive whether TOCP induces hepatotoxicity. The purpose of this study was to investigate the effect of TOCP on hepatocytes and the lipid metabolism in particular. The adult mice were given a single dose of TOCP (800 mg/kg, p.o.) and the histological changes in liver tissue and lipid content in serum were determined. The results showed that more vacuoles and lipid droplets were observed in the liver of the mice exposed to TOCP. And triglyceride concentrations in serum and liver tissue significantly increased. However, the histopathological changes of the liver and the elevated triglyceride levels in the exposed mice can be reversed by endoplasmic reticulum (ER) stress inhibitor 4-phenylbutyric acid and mTOR signal inhibitor rapamycin. It was also found that the changes of expression levels of the biomarkers of ER stress and mTOR signaling pathway, such as GRP78, CHOP, and p-mTOR, in the exposed mice were consistent with those observed in the cultured primary hepatocytes treated with the same chemicals. These results showed that TOCP activated mTOR signal and ER stress to induce de novo lipid synthesis, which led to the hepatic steatosis in mouse.
Collapse
Affiliation(s)
- Jing Li
- Laboratory of Molecular Toxicology, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yi-Jun Wu
- Laboratory of Molecular Toxicology, State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
23
|
Mohibi S, Zhang Y, Perng V, Chen M, Zhang J, Chen X. Ferredoxin 1 is essential for embryonic development and lipid homeostasis. eLife 2024; 13:e91656. [PMID: 38251655 PMCID: PMC10846857 DOI: 10.7554/elife.91656] [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: 08/06/2023] [Accepted: 01/19/2024] [Indexed: 01/23/2024] Open
Abstract
Mammalian ferredoxin 1 and 2 (FDX1/2) belong to an evolutionary conserved family of iron-sulfur cluster containing proteins and act as electron shutters between ferredoxin reductase (FDXR) and numerous proteins involved in critical biological pathways. FDX1 is involved in biogenesis of steroids and bile acids, Vitamin A/D metabolism, and lipoylation of tricarboxylic acid (TCA) cycle enzymes. FDX1 has been extensively characterized biochemically but its role in physiology and lipid metabolism has not been explored. In this study, we generated Fdx1-deficient mice and showed that knockout of both alleles of the Fdx1 gene led to embryonic lethality. We also showed that like Fdxr+/-+/-, Fdx1+/-+/- had a shorter life span and were prone to steatohepatitis. However, unlike Fdxr+/-+/-, Fdx1+/-+/- were not prone to spontaneous tumors. Additionally, we showed that FDX1 deficiency led to lipid droplet accumulation possibly via the ABCA1-SREBP1/2 pathway. Specifically, untargeted lipidomic analysis showed that FDX1 deficiency led to alterations in several classes of lipids, including cholesterol, triacylglycerides, acylcarnitines, ceramides, phospholipids and lysophospholipids. Taken together, our data indicate that FDX1 is essential for mammalian embryonic development and lipid homeostasis at both cellular and organismal levels.
Collapse
Affiliation(s)
- Shakur Mohibi
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, DavisDavisUnited States
| | - Yanhong Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, DavisDavisUnited States
| | - Vivian Perng
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, DavisDavisUnited States
| | - Mingyi Chen
- Department of Pathology, University of Texas Southwestern Medical Center, DallasDallasUnited States
| | - Jin Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, DavisDavisUnited States
| | - Xinbin Chen
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, DavisDavisUnited States
| |
Collapse
|
24
|
Wang X, Chen Y, Meng H, Meng F. SREBPs as the potential target for solving the polypharmacy dilemma. Front Physiol 2024; 14:1272540. [PMID: 38269061 PMCID: PMC10806128 DOI: 10.3389/fphys.2023.1272540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/26/2023] [Indexed: 01/26/2024] Open
Abstract
The phenomenon of polypharmacy is a common occurrence among older people with multiple health conditions due to the rapid increase in population aging and the popularization of clinical guidelines. The prevalence of metabolic syndrome is growing quickly, representing a serious threat to both the public and the worldwide healthcare systems. In addition, it enhances the risk of cardiovascular disease as well as mortality and morbidity. Sterol regulatory element binding proteins (SREBPs) are basic helix-loop-helix leucine zipper transcription factors that transcriptionally modulate genes that regulate lipid biosynthesis and uptake, thereby serving an essential role in biological systems regulation. In this article, we have described the structure of SREBPs and explored their activation and regulation of signals. We also reveal that SREBPs are intricately involved in the modulation of metabolic diseases and thus have tremendous potential as the novel target for single-drug therapy for multiple diseases.
Collapse
Affiliation(s)
| | | | | | - Fanbo Meng
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis (Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute), Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
25
|
Liu H, Chen S, Lin Y, Jiang W, Zhao Y, Lu S, Miao L, Ge X. Ferrous Ion Alleviates Lipid Deposition and Inflammatory Responses Caused by a High Cottonseed Meal Diet by Modulating Hepatic Iron Transport Homeostasis and Controlling Ferroptosis in Juvenile Ctenopharyngodon idellus. Antioxidants (Basel) 2023; 12:1968. [PMID: 38001821 PMCID: PMC10669718 DOI: 10.3390/antiox12111968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
To investigate the mechanisms through which ferrous ion (Fe2+) addition improves the utilization of a cottonseed meal (CSM) diet, two experimental diets with equal nitrogen and energy content (low-cottonseed meal (LCM) and high-cottonseed meal (HCM) diets, respectively) containing 16.31% and 38.46% CSM were prepared. Additionally, the HCM diet was supplemented with graded levels of FeSO4·7H2O to establish two different Fe2+ supplementation groups (HCM + 0.2%Fe2+ and HCM + 0.4%Fe2+). Juvenile Ctenopharyngodon idellus (grass carps) (5.0 ± 0.5 g) were fed one of these four diets (HCM, LCM, HCM + 0.2%Fe2+ and HCM + 0.4%Fe2+ diets) for eight weeks. Our findings revealed that the HCM diet significantly increased lipid peroxide (LPO) concentration and the expression of lipogenic genes, e.g., sterol regulatory element binding transcription factor 1 (srebp1) and stearoyl-CoA desaturase (scd), leading to excessive lipid droplet deposition in the liver (p < 0.05). However, these effects were significantly reduced in the HCM + 0.2%Fe2+ and HCM + 0.4%Fe2+ groups (p < 0.05). Plasma high-density lipoprotein (HDL) concentration was also significantly lower in the HCM and HCM + 0.2%Fe2+ groups compared to the LCM group (p < 0.05), whereas low-density lipoprotein (LDL) concentration was significantly higher in the HCM + 0.2%Fe2+ and HCM + 0.4%Fe2+ groups than in the LCM group (p < 0.05). Furthermore, the plasma levels of liver functional indices, including alkaline phosphatase (ALP), aspartate aminotransferase (AST), alanine aminotransferase (ALT), and glucose (GLU), were significantly lower in the HCM + 0.4%Fe2+ group (p < 0.05). Regarding the expression of genes related to iron transport regulation, transferrin 2 (tfr2) expression in the HCM group and Fe2+ supplementation groups were significantly suppressed compared to the LCM group (p < 0.05). The addition of 0.4% Fe2+ in the HCM diet activated hepcidin expression and suppressed ferroportin-1 (fpn1) expression (p < 0.05). Compared to the LCM group, the expression of genes associated with ferroptosis and inflammation, including acyl-CoA synthetase long-chain family member 4b (acsl4b), lysophosphatidylcholine acyltransferase 3 (lpcat3), cyclooxygenase (cox), interleukin 1β (il-1β), and nuclear factor kappa b (nfκb), were significantly increased in the HCM group (p < 0.05), whereas Fe2+ supplementation in the HCM diet significantly inhibited their expression (p < 0.05) and significantly suppressed lipoxygenase (lox) expression (p < 0.05). Compared with the HCM group without Fe2+ supplementation, Fe2+ supplementation in the HCM diet significantly upregulated the expression of genes associated with ferroptosis, such as heat shock protein beta-associated protein1 (hspbap1), glutamate cysteine ligase (gcl), and glutathione peroxidase 4a (gpx4a) (p < 0.05), and significantly decreased the expression of the inflammation-related genes interleukin 15/10 (il-15/il-10) (p < 0.05). In conclusion, FeSO4·7H2O supplementation in the HCM diet maintained iron transport and homeostasis in the liver of juvenile grass carps, thus reducing the occurrence of ferroptosis and alleviating hepatic lipid deposition and inflammatory responses caused by high dietary CSM contents.
Collapse
Affiliation(s)
- Hengchen Liu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (H.L.); (S.C.); (W.J.); (Y.Z.); (X.G.)
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (Y.L.); (S.L.)
| | - Shiyou Chen
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (H.L.); (S.C.); (W.J.); (Y.Z.); (X.G.)
| | - Yan Lin
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (Y.L.); (S.L.)
| | - Wenqiang Jiang
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (H.L.); (S.C.); (W.J.); (Y.Z.); (X.G.)
| | - Yongfeng Zhao
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (H.L.); (S.C.); (W.J.); (Y.Z.); (X.G.)
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (Y.L.); (S.L.)
| | - Siyue Lu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (Y.L.); (S.L.)
| | - Linghong Miao
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (H.L.); (S.C.); (W.J.); (Y.Z.); (X.G.)
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (Y.L.); (S.L.)
| | - Xianping Ge
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (H.L.); (S.C.); (W.J.); (Y.Z.); (X.G.)
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (Y.L.); (S.L.)
| |
Collapse
|
26
|
Stevenson ER, Smith LC, Wilkinson ML, Lee SJ, Gow AJ. Etiology of lipid-laden macrophages in the lung. Int Immunopharmacol 2023; 123:110719. [PMID: 37595492 PMCID: PMC10734282 DOI: 10.1016/j.intimp.2023.110719] [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: 05/02/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/20/2023]
Abstract
Uniquely positioned as sentinel cells constantly exposed to the environment, pulmonary macrophages are vital for the maintenance of the lung lining. These cells are responsible for the clearance of xenobiotics, pathogen detection and clearance, and homeostatic functions such as surfactant recycling. Among the spectrum of phenotypes that may be expressed by macrophages in the lung, the pulmonary lipid-laden phenotype is less commonly studied in comparison to its circulatory counterpart, the atherosclerotic lesion-associated foam cell, or the acutely activated inflammatory macrophage. Herein, we propose that lipid-laden macrophage formation in the lung is governed by lipid acquisition, storage, metabolism, and export processes. The cellular balance of these four processes is critical to the maintenance of homeostasis and the prevention of aberrant signaling that may contribute to lung pathologies. This review aims to examine mechanisms and signaling pathways that are involved in lipid-laden macrophage formation and the potential consequences of this phenotype in the lung.
Collapse
Affiliation(s)
- E R Stevenson
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States
| | - L C Smith
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States; Department of Pharmaceutical Sciences, University of Connecticut School of Pharmacy, Storrs, CT, United States
| | - M L Wilkinson
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States
| | - S J Lee
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States
| | - A J Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, United States
| |
Collapse
|
27
|
Morishita A, Oura K, Tadokoro T, Fujita K, Tani J, Kobara H, Ono M, Himoto T, Masaki T. MicroRNAs and Nonalcoholic Steatohepatitis: A Review. Int J Mol Sci 2023; 24:14482. [PMID: 37833930 PMCID: PMC10572537 DOI: 10.3390/ijms241914482] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a clinicopathologic syndrome caused by fat deposition in hepatocytes. Patients with nonalcoholic steatohepatitis (NASH), an advanced form of NAFLD with severe fibrosis, are at high risk for liver-related complications, including hepatocellular carcinoma (HCC). However, the mechanism of progression from simple fat deposition to NASH is complex, and previous reports have linked NAFLD to gut microbiota, bile acids, immunity, adipokines, oxidative stress, and genetic or epigenetic factors. NASH-related liver injury involves multiple cell types, and intercellular signaling is thought to be mediated by extracellular vesicles. MicroRNAs (miRNAs) are short, noncoding RNAs that play important roles as post-transcriptional regulators of gene expression and have been implicated in the pathogenesis of various diseases. Recently, many reports have implicated microRNAs in the pathogenesis of NALFD/NASH, suggesting that exosomal miRNAs are potential non-invasive and sensitive biomarkers and that the microRNAs involved in the mechanism of the progression of NASH may be potential therapeutic target molecules. We are interested in which miRNAs are involved in the pathogenesis of NASH and which are potential target molecules for therapy. We summarize targeted miRNAs associated with the etiology and progression of NASH and discuss each miRNA in terms of its pathophysiology, potential therapeutic applications, and efficacy as a NASH biomarker.
Collapse
Affiliation(s)
| | | | - Tomoko Tadokoro
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kita-gun 761-0793, Japan; (A.M.); (K.O.); (K.F.); (J.T.); (H.K.); (M.O.); (T.H.); (T.M.)
| | | | | | | | | | | | | |
Collapse
|
28
|
Shi R, Kernodle SP, Steede TM, Lewis RS. Modified physiology of burley tobacco plants genetically engineered to express Yb 1, a functional EGY enzyme. PLANTA 2023; 258:82. [PMID: 37721629 DOI: 10.1007/s00425-023-04235-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/03/2023] [Indexed: 09/19/2023]
Abstract
MAIN CONCLUSION Transgenic overexpression of a NtEGY2 gene restores normal green color of burley tobacco plants, but does not increase nitrogen utilization efficiency beyond that exhibited by wild-type individuals. Nitrogen physiology is important in tobacco because of its role in generation of leaf yield and accumulation of nitrogen-containing alkaloids that can react with nitrosating agents in the formation of carcinogenic tobacco-specific nitrosamines. Cultivars of the burley tobacco market class are homozygous for deleterious mutant alleles at the duplicate Yb1 and Yb2 loci which have previously been associated with decreased nitrogen use and utilization efficiency; increased leaf nitrate, total nitrogen, and alkaloid levels; and reduced yields. How mutant alleles at these two loci affect these traits is not well understood. Recent characterization of the Yb1 and Yb2 genes (homologs of Arabidopsis EGY1 gene) enabled overexpression of the wild-type Yb1 allele in yb1yb1yb2yb2 plants to determine if observed unfavorable effects were due to linkage or pleiotropy, and to determine if overexpression could lead to beneficial modifications in any of these traits in transgenic plants relative to naturally-occurring wild-type genotypes. Yb1 overexpression was found to confer an agronomic benefit to yb1yb1yb2yb2 genotypes but no advantage to wild-type genotypes. RNA-Seq was used to carry out a comparative transcriptome analysis of genetically engineered and wild-type nearly isogenic lines (NILs) to gain insight on metabolic pathways affecting carbon and nitrogen metabolism that might be altered as the result of genetic variability at the Yb1 and Yb2 loci. Results indicate that complex changes in the transcriptome of tobacco can be manifested by altered expression of Yb1.
Collapse
Affiliation(s)
- Rui Shi
- Department of Crop and Soil Sciences, North Carolina State University, Raleigh, NC, USA
| | - Sheri P Kernodle
- Department of Crop and Soil Sciences, North Carolina State University, Raleigh, NC, USA
| | - Tyler M Steede
- Department of Crop and Soil Sciences, North Carolina State University, Raleigh, NC, USA
| | - Ramsey S Lewis
- Department of Crop and Soil Sciences, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
29
|
Pierdomenico M, Cicero AFG, Veronesi M, Fogacci F, Riccioni C, Benassi B. Effect of Citrus bergamia extract on lipid profile: A combined in vitro and human study. Phytother Res 2023; 37:4185-4195. [PMID: 37312672 DOI: 10.1002/ptr.7897] [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: 09/08/2022] [Revised: 02/28/2023] [Accepted: 05/09/2023] [Indexed: 06/15/2023]
Abstract
With the aim of characterising the hypo-lipidemic function of the Brumex™ ingredient obtained from the whole fruit of Citrus bergamia, a combined pre-clinical and clinical study was conducted. In the HepG2 experimental model, we first demonstrated that Brumex™ does not trigger any significant alteration in cell viability over the tested concentration range of 1-2000 μg/mL (4 and 24 h). By stimulating the phosphorylation of AMP-activated protein kinase (AMPK) at threonine 172, Brumex™ significantly reduces both cholesterol and triglyceride (TG) intracellular content of HepG2 cells and impairs the expression levels of lipid synthesis-related genes (namely, SREBF1c, SREBF2, ACACA, SCD1, HMGCR and FASN). In vitro data have been validated in a dedicated double-blind, placebo-controlled, randomised clinical trial performed in 50 healthy moderately hyper-cholesterolemic subjects, undergoing supplementation with either Brumex™ (400 mg) or placebo for 12 weeks. Clinical and blood laboratory data were evaluated at the baseline and at the end of the trial. Brumex™ positively impacted on both plasma lipid pattern and liver enzymes compared with the placebo, mainly in terms of significant reduction of total cholesterol (TC), TG, low-density lipoprotein-cholesterol (LDL-C), non-high-density lipoprotein-cholesterol (non-HDL-C), apolipoprotein B100 (ApoB), fasting plasma glucose (FPG), glutamic-oxaloacetic transaminase (GOT), glutamate pyruvate transaminase (GPT) and gamma-glutamyl-transferase (gGT).
Collapse
Affiliation(s)
- Maria Pierdomenico
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Rome, Italy
| | - Arrigo F G Cicero
- Hypertension and Cardiovascular Risk Factors Research Center, Medical and Surgical Sciences Deptartment, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Maddalena Veronesi
- Hypertension and Cardiovascular Risk Factors Research Center, Medical and Surgical Sciences Deptartment, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Federica Fogacci
- Hypertension and Cardiovascular Risk Factors Research Center, Medical and Surgical Sciences Deptartment, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | | | - Barbara Benassi
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Rome, Italy
| |
Collapse
|
30
|
Jeon YG, Kim YY, Lee G, Kim JB. Physiological and pathological roles of lipogenesis. Nat Metab 2023; 5:735-759. [PMID: 37142787 DOI: 10.1038/s42255-023-00786-y] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 03/15/2023] [Indexed: 05/06/2023]
Abstract
Lipids are essential metabolites, which function as energy sources, structural components and signalling mediators. Most cells are able to convert carbohydrates into fatty acids, which are often converted into neutral lipids for storage in the form of lipid droplets. Accumulating evidence suggests that lipogenesis plays a crucial role not only in metabolic tissues for systemic energy homoeostasis but also in immune and nervous systems for their proliferation, differentiation and even pathophysiological roles. Thus, excessive or insufficient lipogenesis is closely associated with aberrations in lipid homoeostasis, potentially leading to pathological consequences, such as dyslipidaemia, diabetes, fatty liver, autoimmune diseases, neurodegenerative diseases and cancers. For systemic energy homoeostasis, multiple enzymes involved in lipogenesis are tightly controlled by transcriptional and post-translational modifications. In this Review, we discuss recent findings regarding the regulatory mechanisms, physiological roles and pathological importance of lipogenesis in multiple tissues such as adipose tissue and the liver, as well as the immune and nervous systems. Furthermore, we briefly introduce the therapeutic implications of lipogenesis modulation.
Collapse
Affiliation(s)
- Yong Geun Jeon
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Ye Young Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Gung Lee
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Jae Bum Kim
- Center for Adipocyte Structure and Function, Institute of Molecular Biology and Genetics, School of Biological Sciences, Seoul National University, Seoul, South Korea.
| |
Collapse
|
31
|
Zhong S, Li J, Wei M, Deng Z, Liu X. Fresh and Browned Lotus Root Extracts Promote Cholesterol Metabolism in FFA-Induced HepG2 Cells through Different Pathways. Foods 2023; 12:foods12091781. [PMID: 37174319 PMCID: PMC10178253 DOI: 10.3390/foods12091781] [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: 03/24/2023] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
Browning of fresh-cut plants is mainly attributed to the enzymatic browning of phenolic compounds induced by polyphenol oxidase (PPO), producing browning products such as anthraquinones, flavanol oxides, and glycosides, which are usually considered to be non-toxic. Could browning bring any benefits on behalf of their bioactivity? Our previous study found that browned lotus root extracts (BLREs) could reduce the cholesterol level in obese mice as fresh lotus root extracts (FLREs) did. This study aimed to compare the mechanisms of FLRE and BLRE on cholesterol metabolism and verify whether the main component's monomer regulates cholesterol metabolism like the extracts do through in vitro experiments. Extracts and monomeric compounds are applied to HepG2 cells induced by free fatty acids (FFA). Extracellular total cholesterol (TC) and triglyceride (TG) levels were also detected. In addition, RT-PCR and Western blot were used to observe cholesterol metabolism-related gene and protein expression. The in vitro results showed that BLRE and FLRE could reduce TC and TG levels in HepG2 cells. In addition, BLRE suppressed the synthesis of cholesterol. Meanwhile, FLRE promoted the synthesis of bile acid (BA) as well as the clearance and efflux of cholesterol. Furthermore, the main monomers of BLRE also decreased cholesterol synthesis, which is the same as BLRE. In addition, the main monomers of FLRE promoted the synthesis of BAs, similar to FLRE. BLRE and FLRE promote cholesterol metabolism by different pathways.
Collapse
Affiliation(s)
- Shuyuan Zhong
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Jingfang Li
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Meng Wei
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Xiaoru Liu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| |
Collapse
|
32
|
Hong KU, Walls KM, Hein DW. Non-coding and intergenic genetic variants of human arylamine N-acetyltransferase 2 (NAT2) gene are associated with differential plasma lipid and cholesterol levels and cardiometabolic disorders. Front Pharmacol 2023; 14:1091976. [PMID: 37077812 PMCID: PMC10106703 DOI: 10.3389/fphar.2023.1091976] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/02/2023] [Indexed: 04/05/2023] Open
Abstract
Arylamine N-acetyltransferase 2 (NAT2) is a phase II metabolic enzyme, best known for metabolism of aromatic amines and hydrazines. Genetic variants occurring in the NAT2 coding region have been well-defined and are known to affect the enzyme activity or protein stability. Individuals can be categorized into rapid, intermediate, and slow acetylator phenotypes that significantly alter their ability to metabolize arylamines, including drugs (e.g., isoniazid) and carcinogens (e.g., 4-aminobiphenyl). However, functional studies on non-coding or intergenic variants of NAT2 are lacking. Multiple, independent genome wide association studies (GWAS) have reported that non-coding or intergenic variants of NAT2 are associated with elevated plasma lipid and cholesterol levels, as well as cardiometabolic disorders, suggesting a novel cellular role of NAT2 in lipid and cholesterol homeostasis. The current review highlights and summarizes GWAS reports that are relevant to this association. We also present a new finding that seven, non-coding, intergenic NAT2 variants (i.e., rs4921913, rs4921914, rs4921915, rs146812806, rs35246381, rs35570672, and rs1495741), which have been associated with plasma lipid and cholesterol levels, are in linkage disequilibrium with one another, and thus form a novel haplotype. The dyslipidemia risk alleles of non-coding NAT2 variants are associated with rapid NAT2 acetylator phenotype, suggesting that differential systemic NAT2 activity might be a risk factor for developing dyslipidemia. The current review also discusses the findings of recent reports that are supportive of the role of NAT2 in lipid or cholesterol synthesis and transport. In summary, we review data suggesting that human NAT2 is a novel genetic factor that influences plasma lipid and cholesterol levels and alters the risk of cardiometabolic disorders. The proposed novel role of NAT2 merits further investigations.
Collapse
Affiliation(s)
| | | | - David W. Hein
- Department of Pharmacology & Toxicology, Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, United States
| |
Collapse
|
33
|
Zou C, Tan H, Zeng J, Liu M, Zhang G, Zheng Y, Zhang Z. Hepatitis C virus nonstructural protein 4B induces lipogenesis via the Hippo pathway. Arch Virol 2023; 168:113. [PMID: 36920600 PMCID: PMC10017664 DOI: 10.1007/s00705-023-05743-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/07/2023] [Indexed: 03/16/2023]
Abstract
Hepatitis C virus (HCV) infection causes abnormal lipid metabolism in hepatocytes, which leads to hepatic steatosis and even hepatocellular carcinoma. HCV nonstructural protein 4B (NS4B) has been reported to induce lipogenesis, but the underlying mechanism is unclear. In this study, western blots were performed to investigate the effect of NS4B protein levels on key effectors of the Hippo and AKT signaling pathways. Yes-associated protein (YAP) and moesin-ezrin-radixin-like protein (Merlin) are effectors of the Hippo pathway. NS4B downregulated Merlin and phosphorylated YAP (p-YAP) protein expression while increasing the expression of the key AKT pathway proteins p-AKT and NF-κB. By observing the levels of AKT pathway proteins when Merlin was overexpressed or silenced, it was determined that Merlin mediates the AKT pathway. We suggest that HCV NS4B may mediate the AKT signaling pathway by inhibiting the Hippo pathway. Lipid droplets were observed in Huh7.5 cells overexpressing NS4B, and they increased significantly in number when Merlin was silenced. Overexpression of NS4B and Merlin silencing enhanced the expression of sterol regulatory element binding proteins (SREBPs), which have been demonstrated to be key regulatory factors controlling fatty acid synthesis. NS4B and Merlin silencing also enhanced the in vitro proliferative capacity of hepatocellular carcinoma cells. In conclusion, NS4B induces lipogenesis via the effect of the Hippo-YAP pathway on the AKT signaling pathway and thereby plays a significant role in the pathogenesis of HCV-associated diseases.
Collapse
Affiliation(s)
- Chen Zou
- Department of Pathology, The First Affiliated Hospital, Jinan University, Guangzhou, 510630, China. .,Center for Medical Experiments, University of Chinese Academy of Sciences-Shenzhen Hospital, Shenzhen, 518016, China.
| | - Hongxi Tan
- Center for Medical Experiments, University of Chinese Academy of Sciences-Shenzhen Hospital, Shenzhen, 518016, China
| | - Jun Zeng
- Center for Medical Experiments, University of Chinese Academy of Sciences-Shenzhen Hospital, Shenzhen, 518016, China
| | - Minqi Liu
- Center for Medical Experiments, University of Chinese Academy of Sciences-Shenzhen Hospital, Shenzhen, 518016, China
| | - Guangping Zhang
- Huadu District People's Hospital of Guangzhou, Guangzhou, 510600, China
| | - Yi Zheng
- Center for Medical Experiments, University of Chinese Academy of Sciences-Shenzhen Hospital, Shenzhen, 518016, China
| | - Zhanfeng Zhang
- Department of Laboratory Science, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510600, China.
| |
Collapse
|
34
|
Li Y, Wu S, Zhao X, Hao S, Li F, Wang Y, Liu B, Zhang D, Wang Y, Zhou H. Key events in cancer: Dysregulation of SREBPs. Front Pharmacol 2023; 14:1130747. [PMID: 36969840 PMCID: PMC10030587 DOI: 10.3389/fphar.2023.1130747] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
Lipid metabolism reprogramming is an important hallmark of tumor progression. Cancer cells require high levels of lipid synthesis and uptake not only to support their continued replication, invasion, metastasis, and survival but also to participate in the formation of biological membranes and signaling molecules. Sterol regulatory element binding proteins (SREBPs) are core transcription factors that control lipid metabolism and the expression of important genes for lipid synthesis and uptake. A growing number of studies have shown that SREBPs are significantly upregulated in human cancers and serve as intermediaries providing a mechanistic link between lipid metabolism reprogramming and malignancy. Different subcellular localizations, including endoplasmic reticulum, Golgi, and nucleus, play an indispensable role in regulating the cleavage maturation and activity of SREBPs. In this review, we focus on the relationship between aberrant regulation of SREBPs activity in three organelles and tumor progression. Because blocking the regulation of lipid synthesis by SREBPs has gradually become an important part of tumor therapy, this review also summarizes and analyzes several current mainstream strategies.
Collapse
Affiliation(s)
- Yunkuo Li
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Shouwang Wu
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Xiaodong Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Shiming Hao
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Faping Li
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Yuxiong Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Difei Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
- *Correspondence: Yishu Wang, Honglan Zhou,
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Yishu Wang, Honglan Zhou,
| |
Collapse
|
35
|
Application of Caenorhabditis elegans in Lipid Metabolism Research. Int J Mol Sci 2023; 24:ijms24021173. [PMID: 36674689 PMCID: PMC9860639 DOI: 10.3390/ijms24021173] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/01/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Over the last decade, the development and prevalence of obesity have posed a serious public health risk, which has prompted studies on the regulation of adiposity. With the ease of genetic manipulation, the diversity of the methods for characterizing body fat levels, and the observability of feeding behavior, Caenorhabditis elegans (C. elegans) is considered an excellent model for exploring energy homeostasis and the regulation of the cellular fat storage. In addition, the homology with mammals in the genes related to the lipid metabolism allows many aspects of lipid modulation by the regulators of the central nervous system to be conserved in this ideal model organism. In recent years, as the complex network of genes that maintain an energy balance has been gradually expanded and refined, the regulatory mechanisms of lipid storage have become clearer. Furthermore, the development of methods and devices to assess the lipid levels has become a powerful tool for studies in lipid droplet biology and the regulation of the nematode lipid metabolism. Herein, based on the rapid progress of C. elegans lipid metabolism-related studies, this review outlined the lipid metabolic processes, the major signaling pathways of fat storage regulation, and the primary experimental methods to assess the lipid content in nematodes. Therefore, this model system holds great promise for facilitating the understanding, management, and therapies of human obesity and other metabolism-related diseases.
Collapse
|
36
|
Celik C, Lee SYT, Yap WS, Thibault G. Endoplasmic reticulum stress and lipids in health and diseases. Prog Lipid Res 2023; 89:101198. [PMID: 36379317 DOI: 10.1016/j.plipres.2022.101198] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/14/2022]
Abstract
The endoplasmic reticulum (ER) is a complex and dynamic organelle that regulates many cellular pathways, including protein synthesis, protein quality control, and lipid synthesis. When one or multiple ER roles are dysregulated and saturated, the ER enters a stress state, which, in turn, activates the highly conserved unfolded protein response (UPR). By sensing the accumulation of unfolded proteins or lipid bilayer stress (LBS) at the ER, the UPR triggers pathways to restore ER homeostasis and eventually induces apoptosis if the stress remains unresolved. In recent years, it has emerged that the UPR works intimately with other cellular pathways to maintain lipid homeostasis at the ER, and so does at cellular levels. Lipid distribution, along with lipid anabolism and catabolism, are tightly regulated, in part, by the ER. Dysfunctional and overwhelmed lipid-related pathways, independently or in combination with ER stress, can have reciprocal effects on other cellular functions, contributing to the development of diseases. In this review, we summarize the current understanding of the UPR in response to proteotoxic stress and LBS and the breadth of the functions mitigated by the UPR in different tissues and in the context of diseases.
Collapse
Affiliation(s)
- Cenk Celik
- School of Biological Sciences, Nanyang Technological University, Singapore
| | | | - Wei Sheng Yap
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Guillaume Thibault
- School of Biological Sciences, Nanyang Technological University, Singapore; Mechanobiology Institute, National University of Singapore, Singapore; Institute of Molecular and Cell Biology, A*STAR, Singapore.
| |
Collapse
|
37
|
Ding Q, Zhang G, Wang Y, Xu L, Wu M, Zhou Y, Xu T, Meng X, Huang C, Zhang L. β-catenin ISGylation promotes lipid deposition and apoptosis in ethanol-stimulated liver injury models. Redox Rep 2022; 27:239-248. [PMID: 36259544 PMCID: PMC9586657 DOI: 10.1080/13510002.2022.2109360] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background The restoration of the Wnt/β-catenin pathway to alleviate alcoholic fatty liver disease (AFLD) progression is under study as a new strategy for alcoholic liver disease (ALD) treatment. Recent studies have indicated that interferon-stimulated gene 15 (ISG15) can covalently bind to β-catenin by HECT E3 ubiquitin ligase 5 (HERC5), leading to ISG degradation and downregulation of β-catenin levels. However, the relationship between β-catenin and the ISG15 system in AFLD remains unclear. Methods Here, we explored the roles of the ISG15 system in β-catenin activation and in the pathogenesis of alcohol-induced liver injury and steatosis. Results In this study, HERC5 silencing upregulated β-catenin protein expression and inhibited lipid metabolism disorders and cell apoptosis. Reduced β-catenin protein expression, increased lipid metabolism disorders, and cell apoptosis were detected in cells induced with HERC5 overexpression, which was reversible with the reactive oxygen species (ROS) inhibitor. All the above results were statistically analyzed. Thus, these observations demonstrate that β-catenin ISGylation is a prominent regulator of ALD pathology, which works by regulating ROS to induce lipid metabolism disorders and cell apoptosis. Conclusion Our findings provided the mechanism involved in the β-catenin ISGylation, allowing for future studies on the prevention or amelioration of liver injury in ALD.
Collapse
Affiliation(s)
- Qi Ding
- Anhui No.2 Provincial People's Hospital, Hefei, People's Republic of China
| | - Guodong Zhang
- School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of major autoimmune disease, Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei, People's Republic of China
| | - Yang Wang
- School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of major autoimmune disease, Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei, People's Republic of China
| | - Lei Xu
- School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of major autoimmune disease, Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei, People's Republic of China
| | - Meifei Wu
- School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of major autoimmune disease, Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei, People's Republic of China
| | - Yiwen Zhou
- School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of major autoimmune disease, Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei, People's Republic of China
| | - Tao Xu
- School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of major autoimmune disease, Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei, People's Republic of China
| | - Xiaoming Meng
- School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of major autoimmune disease, Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei, People's Republic of China
| | - Cheng Huang
- School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of major autoimmune disease, Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei, People's Republic of China
| | - Lei Zhang
- School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of major autoimmune disease, Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
38
|
Hong KU, Salazar-González RA, Walls KM, Hein DW. Transcriptional Regulation of Human Arylamine N-Acetyltransferase 2 Gene by Glucose and Insulin in Liver Cancer Cell Lines. Toxicol Sci 2022; 190:158-172. [PMID: 36156098 PMCID: PMC9702998 DOI: 10.1093/toxsci/kfac103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Arylamine N-acetyltransferase 2 (NAT2) is well-known for its role in phase II metabolism of xenobiotics and drugs. More recently, genome wide association studies and murine models implicated NAT2 in regulation of insulin sensitivity and plasma lipid levels. However, the mechanism remains unknown. Transcript levels of human NAT2 varied dynamically in HepG2 (hepatocellular) cells, depending on the nutrient status of the culture media. Culturing the cells in the presence of glucose induced NAT2 mRNA expression as well as its N-acetyltransferase activity significantly. In addition, insulin or acetate treatment also significantly induced NAT2 mRNA. We examined and compared the glucose- and acetate-dependent changes in NAT2 expression to those of genes involved in glucose and lipid metabolism, including FABP1, CPT1A, ACACA, SCD, CD36, FASN, ACLY, G6PC, and PCK1. Genes that are involved in fatty acid transport and lipogenesis, such as FABP1 and CD36, shared a similar pattern of expression with NAT2. In silico analysis of genes co-expressed with NAT2 revealed an enrichment of biological processes involved in lipid and cholesterol biosynthesis and transport. Among these, A1CF (APOBEC1 complementation factor) showed the highest correlation with NAT2 in terms of its expression in normal human tissues. The current study shows, for the first time, that human NAT2 is transcriptionally regulated by glucose and insulin in liver cancer cell lines and that the gene expression pattern of NAT2 is similar to that of genes involved in lipid metabolism and transport.
Collapse
Affiliation(s)
- Kyung U Hong
- Department of Pharmacology & Toxicology and Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| | - Raúl A Salazar-González
- Department of Pharmacology & Toxicology and Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| | - Kennedy M Walls
- Department of Pharmacology & Toxicology and Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| | - David W Hein
- Department of Pharmacology & Toxicology and Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| |
Collapse
|
39
|
Li X, Zhang M, Liu M, Liu TH, Hemba-Waduge RUS, Ji JY. Cdk8 attenuates lipogenesis by inhibiting SREBP-dependent transcription in Drosophila. Dis Model Mech 2022; 15:dmm049650. [PMID: 36305265 PMCID: PMC9702540 DOI: 10.1242/dmm.049650] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 10/14/2022] [Indexed: 10/10/2023] Open
Abstract
Fine-tuning of lipogenic gene expression is important for the maintenance of long-term homeostasis of intracellular lipids. The SREBP family of transcription factors are master regulators that control the transcription of lipogenic and cholesterogenic genes, but the mechanisms modulating SREBP-dependent transcription are still not fully understood. We previously reported that CDK8, a subunit of the transcription co-factor Mediator complex, phosphorylates SREBP at a conserved threonine residue. Here, using Drosophila as a model system, we observed that the phosphodeficient SREBP proteins (SREBP-Thr390Ala) were more stable and more potent in stimulating the expression of lipogenic genes and promoting lipogenesis in vivo than wild-type SREBP. In addition, starvation blocked the effects of wild-type SREBP-induced lipogenic gene transcription, whereas phosphodeficient SREBP was resistant to this effect. Furthermore, our biochemical analyses identified six highly conserved amino acid residues in the N-terminus disordered region of SREBP that are required for its interactions with both Cdk8 and the MED15 subunit of the small Mediator complex. These results support that the concerted actions of Cdk8 and MED15 are essential for the tight regulation of SREBP-dependent transcription. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Xiao Li
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
| | - Meng Zhang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, 1700 Tulane Avenue, New Orleans, LA 70112, USA
| | - Mengmeng Liu
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, 1700 Tulane Avenue, New Orleans, LA 70112, USA
| | - Tzu-Hao Liu
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, 1700 Tulane Avenue, New Orleans, LA 70112, USA
| | - Rajitha-Udakara-Sampath Hemba-Waduge
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, 1700 Tulane Avenue, New Orleans, LA 70112, USA
| | - Jun-Yuan Ji
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, 1700 Tulane Avenue, New Orleans, LA 70112, USA
| |
Collapse
|
40
|
Nezhadebrahimi A, Sepehri H, Jahanshahi M, Marjani M, Marjani A. The effect of simvastatin on gene expression of low-density lipoprotein receptor, sterol regulatory element-binding proteins, stearoyl-CoA desaturase 1 mRNA in rat hepatic tissues. Arch Physiol Biochem 2022; 128:1383-1390. [PMID: 32643419 DOI: 10.1080/13813455.2020.1772829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The study aimed to assess the effect of simvastatin on gene expression of LDLR, SREBPs, and SCD1 in rat hepatic tissues fed with high-fat diets (HFD) and its association with some biochemical parameters. Thirty-two male Wister albino rats were divided into four equal groups (three test and one control groups). The biochemical parameters were determined by using spectrophotometer techniques and the Elisa method. Low-density lipoprotein receptor, sterol regulatory element-binding proteins, stearoyl-CoA desaturase1, Beta-actin were analysed by real-time quantitative polymerase chain reaction (RT-PCR) method. At the end of study, the livers of the rats were separated and changes of hepatic tissue were determined. LDLR, SREBP2, and SCD1 expression increased significantly when compared G1 versus G4 and G2 versus G4. The expression of LDLR, SREBP2, and SCD1 also increased significantly when compared G2 versus G3, G1versus G3 and G1 versus G3 and G2 versus G3. The serum level of cholesterol, triglyceride, glucose, LDL, and HDL increased significantly when compared G1 versus G3. LDL showed significantly decreased when compared G1 versus G2. Cholesterol, glucose and HDL and triglyceride levels were increased significantly when compared G1 versus G4 and G2. Treatment of rats with HFD and simvastatin 20 mg/kg, triglyceride and LDL were almost the same as a control group and LDLR expression increased 98% in liver tissue. Gene expressions may be up-regulated in liver tissue and they showed different effects on biochemical parameters.
Collapse
Affiliation(s)
- Abbas Nezhadebrahimi
- Department of Biochemistry and Biophysics, Student Research Center, Metabolic Disorders Research Center, Gorgan Faculty of Medicine, Golestan University Medical Sciences, Gorgan, Iran
- Department of Physiology, Neuroscience Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Hamid Sepehri
- Department of Physiology, Neuroscience Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehrdad Jahanshahi
- Neuroscience Research Center, Department of Anatomy, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Marjani
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, North Cyprus, Turkey
| | - Abdoljalal Marjani
- Metabolic Disorders Research Center, Department of Biochemistry and Biophysics, Gorgan Faculty of Medicine, Golestan University Medical Sciences, Gorgan, Iran
| |
Collapse
|
41
|
Bengoechea-Alonso MT, Aldaalis A, Ericsson J. Loss of the Fbw7 tumor suppressor rewires cholesterol metabolism in cancer cells leading to activation of the PI3K-AKT signalling axis. Front Oncol 2022; 12:990672. [PMID: 36176395 PMCID: PMC9513553 DOI: 10.3389/fonc.2022.990672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
The sterol regulatory-element binding proteins (SREBPs) are transcription factors controlling cholesterol and fatty acid synthesis and metabolism. There are three SREBP proteins, SREBP1a, SREBP1c and SREBP2, with SREBP1a being the strongest transcription factor. The expression of SREBP1a is restricted to rapidly proliferating cells, including cancer cells. The SREBP proteins are translated as large, inactive precursors bound to the endoplasmic reticulum (ER) membranes. These precursors undergo a two-step cleavage process that releases the amino terminal domains of the proteins, which translocate to the nucleus and function as transcription factors. The nuclear forms of the SREBPs are rapidly degraded by the ubiquitin-proteasome system in a manner dependent on the Fbw7 ubiquitin ligase. Consequently, inactivation of Fbw7 results in the stabilization of active SREBP1 and SREBP2 and enhanced expression of target genes. We report that the inactivation of Fbw7 in cancer cells blocks the proteolytic maturation of SREBP2. The same is true in cells expressing a cancer-specific loss-of-function Fbw7 protein. Interestingly, the activation of SREBP2 is restored in response to cholesterol depletion, suggesting that Fbw7-deficient cells accumulate cholesterol. Importantly, inactivation of SREBP1 in Fbw7-deficient cells also restores the cholesterol-dependent regulation of SREBP2, suggesting that the stabilization of active SREBP1 molecules could be responsible for the blunted activation of SREBP2 in Fbw7-deficient cancer cells. We suggest that this could be an important negative feedback loop in cancer cells with Fbw7 loss-of-function mutations to protect these cells from the accumulation of toxic levels of cholesterol and/or cholesterol metabolites. Surprisingly, we also found that the inactivation of Fbw7 resulted in the activation of AKT. Importantly, the activation of AKT was dependent on SREBP1 and on the accumulation of cholesterol. Thus, we suggest that the loss of Fbw7 rewires lipid metabolism in cancer cells to support cell proliferation and survival.
Collapse
Affiliation(s)
- Maria T. Bengoechea-Alonso
- Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Arwa Aldaalis
- Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Johan Ericsson
- Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
- *Correspondence: Johan Ericsson,
| |
Collapse
|
42
|
Branche E, Wang YT, Viramontes KM, Valls Cuevas JM, Xie J, Ana-Sosa-Batiz F, Shafee N, Duttke SH, McMillan RE, Clark AE, Nguyen MN, Garretson AF, Crames JJ, Spann NJ, Zhu Z, Rich JN, Spector DH, Benner C, Shresta S, Carlin AF. SREBP2-dependent lipid gene transcription enhances the infection of human dendritic cells by Zika virus. Nat Commun 2022; 13:5341. [PMID: 36097162 PMCID: PMC9465152 DOI: 10.1038/s41467-022-33041-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 08/29/2022] [Indexed: 02/07/2023] Open
Abstract
The emergence of Zika virus (ZIKV) as a global health threat has highlighted the unmet need for ZIKV-specific vaccines and antiviral treatments. ZIKV infects dendritic cells (DC), which have pivotal functions in activating innate and adaptive antiviral responses; however, the mechanisms by which DC function is subverted to establish ZIKV infection are unclear. Here we develop a genomics profiling method that enables discrete analysis of ZIKV-infected versus neighboring, uninfected primary human DCs to increase the sensitivity and specificity with which ZIKV-modulated pathways can be identified. The results show that ZIKV infection specifically increases the expression of genes enriched for lipid metabolism-related functions. ZIKV infection also increases the recruitment of sterol regulatory element-binding protein (SREBP) transcription factors to lipid gene promoters, while pharmacologic inhibition or genetic silencing of SREBP2 suppresses ZIKV infection of DCs. Our data thus identify SREBP2-activated transcription as a mechanism for promoting ZIKV infection amenable to therapeutic targeting.
Collapse
Affiliation(s)
- Emilie Branche
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Ying-Ting Wang
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Karla M Viramontes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Joan M Valls Cuevas
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Jialei Xie
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Fernanda Ana-Sosa-Batiz
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Norazizah Shafee
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Sascha H Duttke
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, 99163, USA
| | - Rachel E McMillan
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
- Biomedical Sciences Graduate Program, University of California, La Jolla, CA, 92093, USA
| | - Alex E Clark
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Michael N Nguyen
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Aaron F Garretson
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Jan J Crames
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Nathan J Spann
- Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Zhe Zhu
- Department of Medicine, Division of Regenerative Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
| | - Jeremy N Rich
- Department of Medicine, Division of Regenerative Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Neurology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15232, USA
| | - Deborah H Spector
- Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Christopher Benner
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Sujan Shresta
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA.
| | - Aaron F Carlin
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
43
|
Aldaalis A, Bengoechea-Alonso MT, Ericsson J. The SREBP-dependent regulation of cyclin D1 coordinates cell proliferation and lipid synthesis. Front Oncol 2022; 12:942386. [PMID: 36091143 PMCID: PMC9451027 DOI: 10.3389/fonc.2022.942386] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/25/2022] [Indexed: 12/15/2022] Open
Abstract
The sterol regulatory-element binding protein (SREBP) family of transcription factors regulates cholesterol, fatty acid, and triglyceride synthesis and metabolism. However, they are also targeted by the ubiquitin ligase Fbw7, a major tumor suppressor, suggesting that they could regulate cell growth. Indeed, enhanced lipid synthesis is a hallmark of many human tumors. Thus, the SREBP pathway has recently emerged as a potential target for cancer therapy. We have previously demonstrated that one of these transcription factors, SREBP1, is stabilized and remains associated with target promoters during mitosis, suggesting that the expression of these target genes could be important as cells enter G1 and transcription is restored. Activation of cyclin D-cdk4/6 complexes is critical for the phosphorylation and inactivation of the retinoblastoma protein (Rb) family of transcriptional repressors and progression through the G1 phase of the cell cycle. Importantly, the cyclin D-cdk4/6-Rb regulatory axis is frequently dysregulated in human cancer. In the current manuscript, we demonstrate that SREBP1 activates the expression of cyclin D1, a coactivator of cdk4 and cdk6, by binding to an E-box in the cyclin D1 promoter. Consequently, inactivation of SREBP1 in human liver and breast cancer cell lines reduces the expression of cyclin D1 and attenuates Rb phosphorylation. Rb phosphorylation in these cells can be rescued by restoring cyclin D1 expression. On the other hand, expression of active SREBP1 induced the expression of cyclin D1 and increased the phosphorylation of Rb in a manner dependent on cyclin D1 and cdk4/6 activity. Inactivation of SREBP1 resulted in reduced expression of cyclin D1, attenuated phosphorylation of Rb, and reduced proliferation. Inactivation of SREBP1 also reduced the insulin-dependent regulation of the cyclin D1 gene. At the same time, SREBP1 is known to play an important role in supporting lipid synthesis in cancer cells. Thus, we propose that the SREBP1-dependent regulation of cyclin D1 coordinates cell proliferation with the enhanced lipid synthesis required to support cell growth.
Collapse
Affiliation(s)
- Arwa Aldaalis
- Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Education City, Doha, Qatar
| | - Maria T. Bengoechea-Alonso
- Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Education City, Doha, Qatar
| | - Johan Ericsson
- Division of Biological and Biomedical Sciences, College of Health and Life Sciences, Hamad Bin Khalifa University, Education City, Doha, Qatar
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
- *Correspondence: Johan Ericsson,
| |
Collapse
|
44
|
How CM, Hsiu-Chuan Liao V. Chronic exposure to environmentally relevant levels of di(2-ethylhexyl) phthalate (DEHP) disrupts lipid metabolism associated with SBP-1/SREBP and ER stress in C. elegans. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 307:119579. [PMID: 35671893 DOI: 10.1016/j.envpol.2022.119579] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/24/2022] [Accepted: 06/02/2022] [Indexed: 06/15/2023]
Abstract
DEHP is commonly found in the environment, biota, food, and humans, raising significant health concerns. Whether developmental stage and exposure duration modify the obesogenic effects of DEHP is unclear, especially the underlying mechanisms by which chronic exposure to DEHP as well as its metabolites remain largely unknown. This study investigated the obesogenic effects of chronic DEHP exposure, with levels below environmentally-relevant amounts and provide the mechanism in Caenorhabditis elegans. We show that early-life DEHP exposure resulted in an increased lipid and triglyceride (TG) accumulation mainly attributed to DEHP itself, not its metabolite mono-2-ethylhexyl phthalate (MEHP). In addition, developmental stage and exposure timing influence DEHP-induced TG accumulation and chronic DEHP exposure resulted in the most significant effect. Analysis of fatty acid composition shows that chronic DEHP exposure altered fatty acid composition and TG, resulting in an increased ω-6/ω-3 ratio. The increased TG content by chronic DEHP exposure required lipogenic genes fat-6, fat-7, pod-2, fasn-1, and sbp-1. Moreover, chronic DEHP exposure induced XBP-1-mediated endoplasmic reticulum (ER) stress which might lead to up-regulation of sbp-1. This study suggests the possible involvement of ER stress and SBP-1/SREBP-mediated lipogenesis in chronic DEHP-induced obesogenic effects. Results from this study implies that chronic exposure to DEHP disrupts lipid metabolism, which is likely conserved across species due to evolutionary conservation of molecular mechanisms, raising concerns in ecological and human health.
Collapse
Affiliation(s)
- Chun Ming How
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, 106, Taiwan
| | - Vivian Hsiu-Chuan Liao
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, 106, Taiwan.
| |
Collapse
|
45
|
Osborne TF, Espenshade PJ. Lipid balance must be just right to prevent development of severe liver damage. J Clin Invest 2022; 132:e160326. [PMID: 35642642 PMCID: PMC9151688 DOI: 10.1172/jci160326] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a major health concern that often associates with obesity and diabetes. Fatty liver is usually a benign condition, yet a fraction of individuals progress to severe forms of liver damage, including nonalcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC). Elevated sterol regulatory element-binding protein-driven (SREBP-driven) hepatocyte lipid synthesis is associated with NAFLD in humans and mice. In this issue of the JCI, Kawamura, Matsushita, et al. evaluated the role of SREBP-dependent lipid synthesis in the development of NAFLD, NASH, and HCC in the phosphatase and tensin homolog-knockout (PTEN-knockout) NASH model. Deletion of the gene encoding SREBP cleavage-activating protein (SCAP) from the liver resulted in decreased hepatic lipids, as expected. However, SCAP deletion accelerated progression to more severe liver damage, including NASH and HCC. This study provides a note of caution for those pursuing de novo fat biosynthesis as a therapeutic intervention in human NASH.
Collapse
Affiliation(s)
- Timothy F. Osborne
- Departments of Medicine, Pediatrics, and Biological Chemistry, and Institute for Fundamental Biomedical Research, Johns Hopkins University School of Medicine, St. Petersburg, Florida, USA
| | - Peter J. Espenshade
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
46
|
Dong W, Zhang X, Kong Y, Zhao Z, Mahmoud A, Wu L, Moussian B, Zhang J. CYP311A1 in the anterior midgut is involved in lipid distribution and microvillus integrity in Drosophila melanogaster. Cell Mol Life Sci 2022; 79:261. [PMID: 35478270 PMCID: PMC11072108 DOI: 10.1007/s00018-022-04283-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 04/01/2022] [Indexed: 11/03/2022]
Abstract
Lipids are either taken up from food sources or produced internally in specialized tissues such as the liver. Among others, both routes of lipid metabolism involve cytochrome P450 monooxygenases (CYPs). We sought to analyze the function of Cyp311a1 that has been shown to be expressed in the midgut of the fruit fly Drosophila melanogaster. Using a GFP-tagged version of CYP311A1 that is expressed under the control of its endogenous promoter, we show that Cyp311a1 localizes to the endoplasmic reticulum in epithelial cells of the anterior midgut. In larvae with reduced Cyp311a1 expression in the anterior midgut, compared to control larvae, the apical plasma membrane of the respective epithelial cells contains less and shorter microvilli. In addition, we observed reduction of neutral lipids in the fat body, the insect liver, and decreased phosphatidylethanolamine (PE) and triacylglycerols (TAG) amounts in the whole body of these larvae. Probably as a consequence, they cease to grow and eventually die. The microvillus defects in larvae with reduced Cyp311a1 expression are restored by supplying PE, a major phospholipid of plasma membranes, to the food. Moreover, the growth arrest phenotype of these larvae is partially rescued. Together, these results suggest that the anterior midgut is an import hub in lipid distribution and that the midgut-specific CYP311A1 contributes to this function by participating in shaping microvilli in a PE-dependent manner.
Collapse
Affiliation(s)
- Wei Dong
- Research Institute of Applied Biology, Shanxi University, Taiyuan, 030006, Shanxi, China
| | - Xubo Zhang
- Research Institute of Applied Biology, Shanxi University, Taiyuan, 030006, Shanxi, China
| | - Yue Kong
- Research Institute of Applied Biology, Shanxi University, Taiyuan, 030006, Shanxi, China
| | - Zhenwen Zhao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, China
| | - Ali Mahmoud
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse-108, 01307, Dresden, Germany
| | - Lixian Wu
- Research Institute of Applied Biology, Shanxi University, Taiyuan, 030006, Shanxi, China
| | - Bernard Moussian
- Université Côte d'Azur, Parc Valrose, 06108, Nice Cedex 2, France.
| | - Jianzhen Zhang
- Research Institute of Applied Biology, Shanxi University, Taiyuan, 030006, Shanxi, China.
| |
Collapse
|
47
|
Yu Z, Feng Z, Fu L, Wang J, Li C, Zhu H, Xie T, Zhou J, Zhou L, Zhou X. Qingluotongbi formula regulates the LXRα-ERS-SREBP-1c pathway in hepatocytes to alleviate the liver injury caused by Tripterygium wilfordii Hook. f. JOURNAL OF ETHNOPHARMACOLOGY 2022; 287:114952. [PMID: 34968661 DOI: 10.1016/j.jep.2021.114952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 12/04/2021] [Accepted: 12/23/2021] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tripterygium wilfordii Hook. f. (TW) is widely used to treat autoimmune and inflammatory diseases; however, its development and application is limited by its significant association with liver injury. The compound formula Qingluotongbi (QLT) employs TW as its main component and is used to treat rheumatoid arthritis with no adverse reactions, suggesting that QLT may reduce the liver toxicity of TW. AIM OF THE STUDY We examined whether TW interferes with lipid metabolism to induce liver injury, and evaluated the protective effect of QLT in in vivo and in vitro experiments. MATERIALS AND METHODS After administration of QLT and its ingredients, HepaRG cells and SD rats were tested for biochemical indicators, hepatocytes lipid changes, and rat liver pathological changes, and then we analyzed for the gene expression of liver X receptor α (LXRα), endoplasmic reticulum stress (ERS) key proteins, sterol regulatory element binding protein-1c (SREBP-1c), and lipid-synthesizing enzymes. In HepaRG cells, the protein expression of glucose-regulated protein 78 kDa (GRP78) and LXRα was detected after addition of an LXRα inhibitor, LXRα agonist, and ERS inhibitor. RESULTS TW caused significant elevation of biochemical indicators and lipid droplet deposition in hepatocytes, as well as upregulated the gene expression of LXRα, ERS key proteins, SREBP-1c, and lipid-synthesizing enzymes in both in vitro and in vivo settings, and caused liver injury in rats. QLT can alleviate the lipotoxic liver injury caused by TW. LXRα agonist further activated ERS induced by TW, whereas LXRα inhibitor significantly reduced ERS and lipotoxic injury induced by TW in HepaRG cells. CONCLUSIONS TW upregulated LXRα to activate ERS and increased the gene expression of SREBP-1c and lipid-synthesizing enzymes, leading to increased lipid synthesis in hepatocytes to result in liver injury. QLT inhibited the LXRα-ERS-SREBP-1c pathway and reduced abnormal lipid synthesis in hepatocytes and the hepatotoxicity of TW.
Collapse
Affiliation(s)
- Zhichao Yu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Zhe Feng
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Ling Fu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Jing Wang
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Changqing Li
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Huaxu Zhu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources, Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Tong Xie
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Jie Zhou
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Lingling Zhou
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Xueping Zhou
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| |
Collapse
|
48
|
Zhang Y, Mohibi S, Vasilatis DM, Chen M, Zhang J, Chen X. Ferredoxin reductase and p53 are necessary for lipid homeostasis and tumor suppression through the ABCA1-SREBP pathway. Oncogene 2022; 41:1718-1726. [PMID: 35121827 PMCID: PMC8933276 DOI: 10.1038/s41388-021-02100-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 10/18/2021] [Accepted: 10/25/2021] [Indexed: 12/21/2022]
Abstract
p53 is known to modulate metabolism and FDXR is required for steroidogenesis. Given that FDXR is a target/regulator of p53, the FDXR–p53 axis may play a unique role in lipid metabolism. Here, we found that expression of ABCA1, a cholesterol-efflux pump, was suppressed by loss of FDXR and/or p53, leading to activation of master lipogenic regulators SREBP1/2. Accordingly, lipid droplets, cholesterol, and triglycerides were increased by loss of FDXR or p53, which were further increased by loss of both FDXR and p53. To explore the biological significance of the FDXR–p53 axis, we generated a cohort of mice deficient in Fdxr and/or Trp53. We found that Fdxr+/−, Trp53+/−, and Fdxr+/−;Trp53+/− mice had a short life span and were prone to spontaneous tumors and liver steatosis. Moreover, the levels of serum cholesterol and triglycerides were significantly increased in Fdxr+/− and Trp53+/− mice, which were further increased in Fdxr+/−;Trp53+/− mice. Interestingly, loss of Fdxr but not p53 led to accumulation of serum low-density lipoprotein. Together, our findings reveal that the FDXR–p53 axis plays a critical role in lipid homeostasis and tumor suppression.
Collapse
Affiliation(s)
- Yanhong Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Shakur Mohibi
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Demitria M Vasilatis
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, CA, 95616, USA
| | - Mingyi Chen
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jin Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, CA, 95616, USA.
| | - Xinbin Chen
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California at Davis, Davis, CA, 95616, USA.
| |
Collapse
|
49
|
Hirose S, Asano K, Harada S, Takahashi T, Kondou E, Ito K, Iddamalgoda A, Nakane A. Effects of salmon cartilage proteoglycan on obesity in mice fed with a high-fat diet. Food Sci Nutr 2022; 10:577-583. [PMID: 35154693 PMCID: PMC8825722 DOI: 10.1002/fsn3.2685] [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: 07/01/2021] [Revised: 11/09/2021] [Accepted: 11/14/2021] [Indexed: 11/05/2022] Open
Abstract
This study investigated the effects of salmon nasal cartilage proteoglycan (PG), which shows anti-inflammatory properties, on obesity induced by high-fat diet (HFD) in a mouse model. Mice were fed either a HFD or normal diet (ND), with or without PG, for 8-12 weeks. After 12 weeks, the body weight of mice fed with PG-free HFD was 54.08 ± 4.67 g, whereas that of mice fed with HFD containing PG was 41.83 ± 4.97 g. The results suggest that the increase in body weight was attenuated in mice fed with HFD containing PG. This effect was not observed in mice fed with ND. The PG administration suppressed the elevation of serum lipids (the level of serum lipids ranged between 54% and 69% compared to 100% in mice fed with PG-free HFD) and the upregulated mRNA expression of sterol regulatory element-binding protein-1c (SREBP-1c), which is a transcription factor that acts as a master regulator of lipogenic gene expression in the liver (the expression level was 77.5% compared to 100% in mice fed with PG-free HFD). High leptin levels in mice fed with PG-free HFD were observed during fasting (average at 14,376 ng/ml), and they did not increase after refeeding (average of 14,263 ng/ml), whereas serum leptin levels in mice fed with HFD containing PG were low during fasting (average of 6481 ng/ml) and increased after refeeding (average 13,382 ng/ml). These results suggest that PG feeding has an anti-obesity effect and that the regulation of SREBP-1c and leptin secretion play a role in this effect.
Collapse
Affiliation(s)
- Shouhei Hirose
- Department of Biopolymer and Health ScienceHirosaki University Graduate School of MedicineHirosakiJapan
- Department of Microbiology and ImmunologyHirosaki University Graduate School of MedicineHirosakiJapan
- Present address:
Division of MicrobiologyNational Institute of Health SciencesKawasakiJapan
| | - Krisana Asano
- Department of Biopolymer and Health ScienceHirosaki University Graduate School of MedicineHirosakiJapan
- Department of Microbiology and ImmunologyHirosaki University Graduate School of MedicineHirosakiJapan
| | - Seiyu Harada
- Department of HealthcareDydo DRINCO, Inc.OsakaJapan
| | - Tatsuji Takahashi
- Department of Biopolymer and Health ScienceHirosaki University Graduate School of MedicineHirosakiJapan
- Department of Research and DevelopmentIchimaru Pharcos Co., Ltd.Motosu CityJapan
| | - Eriko Kondou
- Department of HealthcareDydo DRINCO, Inc.OsakaJapan
| | - Kenichi Ito
- Department of Research and DevelopmentIchimaru Pharcos Co., Ltd.Motosu CityJapan
| | | | - Akio Nakane
- Department of Biopolymer and Health ScienceHirosaki University Graduate School of MedicineHirosakiJapan
- Department of Nursing and School of Health SciencesHirosaki University of Health and WelfareHirosakiJapan
| |
Collapse
|
50
|
Abstract
Interleukin 17A (IL-17A)-producing T helper 17 (Th17) cells were identified as a subset of T helper cells that play a critical role in host defense against bacterial and fungal pathogens. Th17 cells differentiate from Th0 naïve T-cells in response to transforming growth factor β1 (TGF-β1) and IL-6, the cytokines which also drive development of liver fibrosis, require activation of transcription factor retinoic acid receptor-related orphan nuclear receptor gamma t (RORγt). IL-17A signals through the ubiquitously expressed receptor IL-17RA. Expression of IL-17RA is upregulated in patients with hepatitis B virus/hepatitis C virus (HBV/HCV) infections, nonalcoholic steatohepatitis (NASH), alcohol-associated liver disease (AALD), hepatocellular carcinoma (HCC), and experimental models of chronic toxic liver injury. The role of IL-17 signaling in the pathogenesis of NASH- and AALD-induced metabolic liver injury and HCC will be the focus of this review. The role of IL-17A-IL-17RA axis in mediation of the cross-talk between metabolically injured hepatic macrophages, hepatocytes, and fibrogenic myofibroblasts will be discussed.
Collapse
Affiliation(s)
- Na Li
- Shanghai University of Medicine & Health Sciences, Shanghai, P.R. China.,Department of Medicine, University of California, San Diego, La Jolla, CA.,Department of Surgery, University of California, San Diego, La Jolla, CA
| | - Gen Yamamoto
- Department of Medicine, University of California, San Diego, La Jolla, CA.,Department of Surgery, University of California, San Diego, La Jolla, CA
| | - Hiroaki Fuji
- Department of Medicine, University of California, San Diego, La Jolla, CA.,Department of Surgery, University of California, San Diego, La Jolla, CA
| | - Tatiana Kisseleva
- Department of Surgery, University of California, San Diego, La Jolla, CA
| |
Collapse
|