1
|
Luse MA, Schug WJ, Dunaway LS, Nyshadham S, Loeb SA, Carvalho A, Tessema R, Pavelic C, Keller TCS, Shu X, Ruddiman CA, Kosmach A, Sveeggen TM, Mitchell R, Bagher P, Minshal RD, Leitnger N, Columbus L, Levental KR, Levental I, Cortese-Krott M, Isakson BE. Nitrosation of CD36 Regulates Endothelial Function and Serum Lipids. Arterioscler Thromb Vasc Biol 2025. [PMID: 40242868 DOI: 10.1161/atvbaha.124.321964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND During obesity, endothelial cells (ECs) become lipid laden, leading to endothelial dysfunction. We tested posttranslational modification on CD36 that may regulate EC lipid accumulation. METHODS We used an EC-specific Cav1 (caveolin-1) knockout mouse, nitrosation and palmitoylation assays, and whole animal Nγ-nitro-l-arginine methyl ester administration to examine blood lipids. RESULTS EC-specific Cav1 knockout male mice are hyperlipidemic regardless of diet but retain endothelial cell function. We found these mice have significantly increased NO in response to the lack of Cav1, and the presence or absence of NO toggled inversely EC lipid content and plasma lipid in mice. The NO nitrosated the fatty acid translocase CD36 at the same cysteines that are palmitoylated on CD36. The nitrosation of CD36 prevented its trafficking to the plasma membrane and decreased lipid accumulation. The physiological effect of this mechanism was a reliance on NO for endothelial function and not dilation. CONCLUSIONS This work suggests that CD36 nitrosation occurs as a protective mechanism to prevent EC lipotoxicity.
Collapse
Affiliation(s)
- Melissa A Luse
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| | - Wyatt J Schug
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| | - Luke S Dunaway
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
| | - Shruthi Nyshadham
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
| | - Skylar A Loeb
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| | - Alicia Carvalho
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
| | - Rachel Tessema
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
| | - Caitlin Pavelic
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville (C.P., C.A.R., N.L.)
| | - T C Stevenson Keller
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| | - Xiaohong Shu
- College of Pharmacy, Dalian Medical University, China (X.S.)
| | - Claire A Ruddiman
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville (C.P., C.A.R., N.L.)
| | - Anna Kosmach
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (A.K., T.M.S., R.M., P.B.)
| | - Timothy M Sveeggen
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (A.K., T.M.S., R.M., P.B.)
| | - Ray Mitchell
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (A.K., T.M.S., R.M., P.B.)
| | - Pooneh Bagher
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha (A.K., T.M.S., R.M., P.B.)
| | | | - Norbert Leitnger
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville (C.P., C.A.R., N.L.)
| | - Linda Columbus
- Department of Chemistry, University of Virginia, Charlottesville (L.C.)
| | - Kandice R Levental
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| | - Ilya Levental
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| | | | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center (M.A.L., W.J.S., L.S.D., S.N., S.A.L., A.C., R.T., C.P., T.C.S.K., C.A.R., N.L., B.E.I.)
- Department of Molecular Physiology and Biophysics (M.A.L., W.J.S., S.A.L., T.C.S.K., K.R.L., I.L., B.E.I.)
| |
Collapse
|
2
|
Xu H, Li Y, Guo N, Wu S, Liu C, Gui Z, Xue W, Jiang X, Ye M, Geng Q, Feng X, Zhang C, Jin L, Hu C. Caveolin-1 mitigates the advancement of metabolic dysfunction-associated steatotic liver disease by reducing endoplasmic reticulum stress and pyroptosis through the restoration of cholesterol homeostasis. Int J Biol Sci 2025; 21:490-506. [PMID: 39781461 PMCID: PMC11705642 DOI: 10.7150/ijbs.100794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/19/2024] [Indexed: 01/12/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent chronic liver disease worldwide, which has the potential to advance to fibrosis. CAV1 has the effects of improving liver lipid deposition in MASLD, however, the potential mechanism is largely unknown. Here, we establish a MASLD mouse model in CAV1 knockout (KO) mice and perform transcriptome analysis on livers from mice to investigate the effects of CAV1 in MASLD progression. In addition, we evaluated the expression of CAV1 in human liver samples, and also conducted assays in vitro to investigate the molecular role of CAV1 in MASLD progression. The results illustrate that the expression of liver CAV1 in the decreases during MASLD progression, which aggravates the accumulation of cholesterol in the liver, leading to more severe endoplasmic reticulum (ER) stress and pyroptosis. Mechanistically, CAV1 regulates the expression of FXR/NR1H4 and its downstream cholesterol transporter, ABCG5/ABCG8, suppressing ER stress and alleviating pyroptosis. Our study confirms CAV1 is a crucial regulator of cholesterol homeostasis in MASLD and plays an important role in disease progression.
Collapse
Affiliation(s)
- Hanlin Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Yu Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Ning Guo
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Shuai Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Can Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhongxuan Gui
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Weiju Xue
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Xiangfu Jiang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Mengjia Ye
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Qianqian Geng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Xiaowen Feng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Chao Zhang
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Lei Jin
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chengmu Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, China
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| |
Collapse
|
3
|
Wu S, Guo N, Xu H, Li Y, Sun T, Jiang X, Fu D, You T, Diao S, Huang Y, Hu C. Caveolin-1 ameliorates hepatic injury in non-alcoholic fatty liver disease by inhibiting ferroptosis via the NOX4/ROS/GPX4 pathway. Biochem Pharmacol 2024; 230:116594. [PMID: 39490677 DOI: 10.1016/j.bcp.2024.116594] [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: 05/04/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease globally, with a complex and contentious pathogenesis. Caveolin-1 (CAV1) is an important regulator of liver function and can mitigate liver injury by scavenging reactive oxygen species (ROS). Evidence suggests that NOX4 is a source of ROS production, that oxidative stress and ferroptosis are closely related, and that both are involved in the onset and progression of NAFLD. However, whether CAV1 attenuates liver injury in NAFLD caused by high-fat diet via the NOX4/ROS/GPX4 pathway remains unclear. An in vivo fatty liver model was established by feeding mice with a high-fat diet for 16 weeks. In addition, an in vitro fatty liver model was established by incubating AML-12 cells with free fatty acids for 24 h using an in vitro culture method. In our study, it was observed that a high-fat diet induces mitochondrial damage and worsens oxidative stress in NAFLD. This diet also hinders GPX4 expression, leading to an escalation of ferroptosis and lipid accumulation. To counteract these effects, intraperitoneal administration of CSD peptide in mice attenuated the high-fat diet-induced liver mitochondrial damage and ferroptosis. Likewise, overexpression of CAV1 resulted in an increase in GPX4 expression and a reduction in levels of ROS-mediated iron metamorphosis, thus mitigating the progression of the disease. However, the effects of CAV1 on GPX4-mediated ferroptosis and lipid deposition could be reversed by CAV1 small interfering RNA (SiRNA). Finally, NOX4 inhibitor (GLX351322) treatment increased CAV1 siRNA-mediated GPX4 expression and decreased the level of ROS-mediated ferroptosis. These findings suggest a potential mechanism underlying the protective role of CAV1 against high-fat diet-induced hepatotoxicity in NAFLD, shedding new light on the interplay between CAV1, GPX4, and ferroptosis in liver pathology.
Collapse
Affiliation(s)
- Shuai Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Ning Guo
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Hanlin Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Yu Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Tianyin Sun
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Xiangfu Jiang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Dongdong Fu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Tingyu You
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Shaoxi Diao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Yan Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Chengmu Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
4
|
Feng X, Zhang R, Yang Z, Zhang K, Xing J. Mechanism of Metabolic Dysfunction-associated Steatotic Liver Disease: Important role of lipid metabolism. J Clin Transl Hepatol 2024; 12:815-826. [PMID: 39280069 PMCID: PMC11393839 DOI: 10.14218/jcth.2024.00019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 08/02/2024] [Accepted: 08/08/2024] [Indexed: 09/18/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease, has a high global prevalence and can progress to metabolic dysfunction-associated steatohepatitis, cirrhosis, and hepatocellular carcinoma. The pathogenesis of MASLD is primarily driven by disturbances in hepatic lipid metabolism, involving six key processes: increased hepatic fatty acid uptake, enhanced fatty acid synthesis, reduced oxidative degradation of fatty acids, increased cholesterol uptake, elevated cholesterol synthesis, and increased bile acid synthesis. Consequently, maintaining hepatic lipid metabolic homeostasis is essential for effective MASLD management. Numerous novel molecules and Chinese proprietary medicines have demonstrated promising therapeutic potential in treating MASLD, primarily by inhibiting lipid synthesis and promoting lipid oxidation. In this review, we summarized recent research on MASLD, elucidated the molecular mechanisms by which lipid metabolism disorders contribute to MASLD pathogenesis, and discussed various lipid metabolism-targeted therapeutic approaches for MASLD.
Collapse
Affiliation(s)
- Xiaoxi Feng
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Rutong Zhang
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhenye Yang
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Kaiguang Zhang
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jun Xing
- Department of Digestive Disease, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
5
|
Rein-Fischboeck L, Pohl R, Haberl EM, Mages W, Girke P, Liebisch G, Krautbauer S, Buechler C. Lower adiposity does not protect beta-2 syntrophin null mice from hepatic steatosis and inflammation in experimental non-alcoholic steatohepatitis. Gene 2023; 859:147209. [PMID: 36681100 DOI: 10.1016/j.gene.2023.147209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 12/21/2022] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
Visceral adiposity is strongly associated with liver steatosis, which predisposes to the development of non-alcoholic steatohepatitis (NASH). Mice with loss of the molecular adapter protein beta-2 syntrophin (SNTB2) have greatly reduced intra-abdominal fat mass. Hepatic expression of proteins with a role in fatty acid metabolism such as fatty acid synthase was nevertheless normal. This was also the case for proteins regulating cholesterol synthesis and uptake. Yet, a slight induction of hepatic cholesterol was noticed in the mutant mice. When mice were fed a methionine choline deficient (MCD) diet to induce NASH, liver cholesteryl ester content was induced in the wild type but not the mutant mice. Serum cholesterol of the mice fed a MCD diet declined and this was significant for the SNTB2 null mice. Though the mutant mice lost less fat mass than the wild type animals, hepatic triglyceride levels were similar between the groups. Proteins involved in fatty acid or cholesterol metabolism such as fatty acid synthase, apolipoprotein E and low-density lipoprotein receptor did not differ between the genotypes. Hepatic oxidative stress and liver inflammation of mutant and wild type mice were comparable. Mutant mice had lower hepatic levels of secondary bile acids and higher cholesterol storage in epididymal fat, and this may partly prevent hepatic cholesterol deposition. In summary, the current study shows that SNTB2 null mice have low intra-abdominal fat mass and do not accumulate hepatic cholesteryl esters when fed a MCD diet. Nevertheless, the SNTB2 null mice develop a similar NASH pathology as wild type mice suggesting a minor role of intra-abdominal fat and liver cholesteryl esters in this model.
Collapse
Affiliation(s)
- Lisa Rein-Fischboeck
- Department of Internal Medicine I, Regensburg University Hospital, D-93053 Regensburg, Germany
| | - Rebekka Pohl
- Department of Internal Medicine I, Regensburg University Hospital, D-93053 Regensburg, Germany
| | - Elisabeth M Haberl
- Department of Internal Medicine I, Regensburg University Hospital, D-93053 Regensburg, Germany
| | - Wolfgang Mages
- Department of Genetics, University of Regensburg, D-93040 Regensburg, Germany
| | - Philipp Girke
- Department of Genetics, University of Regensburg, D-93040 Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, D-93053 Regensburg, Germany
| | - Sabrina Krautbauer
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, D-93053 Regensburg, Germany
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, D-93053 Regensburg, Germany.
| |
Collapse
|
6
|
Diclofenac Disrupts the Circadian Clock and through Complex Cross-Talks Aggravates Immune-Mediated Liver Injury-A Repeated Dose Study in Minipigs for 28 Days. Int J Mol Sci 2023; 24:ijms24021445. [PMID: 36674967 PMCID: PMC9863319 DOI: 10.3390/ijms24021445] [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: 11/22/2022] [Revised: 12/28/2022] [Accepted: 12/30/2022] [Indexed: 01/14/2023] Open
Abstract
Diclofenac effectively reduces pain and inflammation; however, its use is associated with hepato- and nephrotoxicity. To delineate mechanisms of injury, we investigated a clinically relevant (3 mg/kg) and high-dose (15 mg/kg) in minipigs for 4 weeks. Initially, serum biochemistries and blood-smears indicated an inflammatory response but returned to normal after 4 weeks of treatment. Notwithstanding, histopathology revealed drug-induced hepatitis, marked glycogen depletion, necrosis and steatosis. Strikingly, the genomic study revealed diclofenac to desynchronize the liver clock with manifest inductions of its components CLOCK, NPAS2 and BMAL1. The > 4-fold induced CRY1 expression underscored an activated core-loop, and the dose dependent > 60% reduction in PER2mRNA repressed the negative feedback loop; however, it exacerbated hepatotoxicity. Bioinformatics enabled the construction of gene-regulatory networks, and we linked the disruption of the liver-clock to impaired glycogenesis, lipid metabolism and the control of immune responses, as shown by the 3-, 6- and 8-fold induced expression of pro-inflammatory CXCL2, lysozyme and ß-defensin. Additionally, diclofenac treatment caused adrenocortical hypertrophy and thymic atrophy, and we evidenced induced glucocorticoid receptor (GR) activity by immunohistochemistry. Given that REV-ERB connects the circadian clock with hepatic GR, its > 80% repression alleviated immune responses as manifested by repressed expressions of CXCL9(90%), CCL8(60%) and RSAD2(70%). Together, we propose a circuitry, whereby diclofenac desynchronizes the liver clock in the control of the hepatic metabolism and immune response.
Collapse
|
7
|
Qin T, Liu M, Lv Y, Zheng A, Wang L, Wu Y, Kasianenko O, Wei X, Teng Z, Xia X, Hu J. Comprehensive Analysis of lncRNA and mRNA Expression Profile of Macrophage RAW264.7 Stimulated by Antimicrobial Peptide BSN-37. Protein Pept Lett 2023; 30:783-793. [PMID: 37587823 DOI: 10.2174/0929866530666230816110009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/16/2023] [Accepted: 07/06/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND BSN-37, a novel antimicrobial peptide (AMP) containing 37 amino acid residues isolated from the bovine spleen, has not only antibacterial activity but also immunomodulatory activity. Recent evidence shows that long non-coding RNAs (lncRNAs) play an important role in regulating the activation and function of immune cells. The purpose of this experiment was to investigate the lncRNA and mRNA expression profile of mouse macrophages RAW264.7 stimulated by bovine antimicrobial peptide BSN-37. METHODS The whole gene expression microarray was used to detect the differentially expressed lncRNA and mRNA between antimicrobial peptide BSN-37 activated RAW264.7 cells and normal RAW264.7 cells. KEGG pathway analysis and GO function annotation analysis of differentially expressed lncRNAs and mRNA were carried out. Eight kinds of lncRNAs and nine kinds of mRNA with large differences were selected for qRT-PCR verification, respectively. RESULTS In the current study, we found that 1294 lncRNAs and 260 mRNAs were differentially expressed between antibacterial peptide BSN-37 treatment and control groups. Among them, Bcl2l12, Rab44, C1s, Cd101 and other genes were associated with immune responses and were all significantly up-regulated. Mest and Prkcz are related to cell growth, and other genes are related to glucose metabolism and lipid metabolism. In addition, some immune-related terms were also found in the GO and KEGG analyses. At the same time, real-time quantitative PCR was used to verify selected lncRNA and mRNA with differential expression. The results of qRT-PCR verification were consistent with the sequencing results, indicating that our data were reliable. CONCLUSION This study provides the lncRNA and mRNA expression profiles of RAW264.7 macrophages stimulated by antimicrobial peptide BSN-37 and helps to provide a reference value for subsequent studies on lncRNA regulation of antimicrobial peptide BSN-37 immune function.
Collapse
Affiliation(s)
- Ting Qin
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Mingcheng Liu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
- Faculty of Veterinary Medicine, Sumy National Agrarian University, Sumy, Ukraine
| | - Yanhe Lv
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Airong Zheng
- Forage and Feed Station of Henan Province, Zhengzhou, China
| | - Lei Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Yundi Wu
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, China
| | - Oksana Kasianenko
- Faculty of Veterinary Medicine, Sumy National Agrarian University, Sumy, Ukraine
| | - Xiaobing Wei
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Zhanwei Teng
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Xiaojing Xia
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| | - Jianhe Hu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, China
| |
Collapse
|
8
|
Li S, Chen H, Jiang X, Hu F, Li Y, Xu G. Adeno-associated virus-based caveolin-1 delivery via different routes for the prevention of cholesterol gallstone formation. Lipids Health Dis 2022; 21:109. [PMID: 36303150 PMCID: PMC9609467 DOI: 10.1186/s12944-022-01718-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hepatic caveolin-1 (CAV1) is reduced in cholesterol gallstone disease (CGD). Mice with CAV1 deficiency were prone to develop CGD. However, it remains unknown whether restored hepatic CAV1 expression prevents the development of CGD. METHODS C57BL/6 mice were injected with adeno-associated virus 2/8 (AAV2/8) vectors carrying the CAV1 gene (AAV2/8CAV1) via intravenous (i.v.) or intraperitoneal (i.p.) route and then subjected to a lithogenic diet (LD) for 8 weeks. Uninjected mice were used as controls. The functional consequences of rescuing CAV1 expression by either i.v. or i.p. AAV2/8CAV1 treatment for CGD prevention and its subsequent molecular mechanisms were examined. RESULTS CAV1 expression was reduced in the liver and gallbladder of LD-fed CGD mice. We discovered that AAV2/8CAV1 i.p. delivery results in higher transduction efficiency in the gallbladder than tail vein administration. Although either i.v. or i.p. injection of AAV2/8CAV1 improved liver lipid metabolic abnormalities in CGD mice but did not affect LD feeding-induced bile cholesterol supersaturation. In comparison with i.v. administration route, i.p. administration of AAV2/8CAV1 obviously increased CAV1 protein levels in the gallbladder of LD-fed mice, and i.p. delivery of AAV2/8CAV1 partially improved gallbladder cholecystokinin receptor (CCKAR) responsiveness and impeded bile cholesterol nucleation via the activation of adenosine monophosphate-activated protein kinase (AMPK) signaling, which induced a reduction in gallbladder mucin-1 (MUC1) and MUC5ac expression and gallbladder cholesterol accumulation. CONCLUSION CGD prevention by i.p. AAV2/8CAV1 injection in LD-fed mice was associated with the improvement of gallbladder stasis, which again supported the notion that supersaturated bile is required but not sufficient for the formation of cholesterol gallstones. Additionally, AAV treatment via the local i.p. injection offers particular advantages over the systemic i.v. route for much more effective gallbladder gene delivery, which will be an excellent tool for conducting preclinical functional studies on the maintenance of normal gallbladder function to prevent CGD.
Collapse
Affiliation(s)
- Sha Li
- grid.13402.340000 0004 1759 700XDepartment of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, 310006 Hangzhou, Zhejiang China
| | - Hongtan Chen
- grid.13402.340000 0004 1759 700XDepartment of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, 310006 Hangzhou, Zhejiang China
| | - Xin Jiang
- grid.13402.340000 0004 1759 700XDepartment of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, 310006 Hangzhou, Zhejiang China
| | - Fengling Hu
- grid.13402.340000 0004 1759 700XDepartment of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, 310006 Hangzhou, Zhejiang China
| | - Yiqiao Li
- grid.417401.70000 0004 1798 6507Urology & Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital and Hangzhou Medical College Affiliated People’s Hospital, 158 Shangtang Road, 310014 Hangzhou, Zhejiang China
| | - Guoqiang Xu
- grid.13402.340000 0004 1759 700XDepartment of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, 310006 Hangzhou, Zhejiang China
| |
Collapse
|
9
|
Young EN, Dogan M, Watkins C, Bajwa A, Eason JD, Kuscu C, Kuscu C. A Review of Defatting Strategies for Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2022; 23:ijms231911805. [PMID: 36233107 PMCID: PMC9569609 DOI: 10.3390/ijms231911805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/30/2022] Open
Abstract
Non-alcoholic fatty liver disease is a huge cause of chronic liver failure around the world. This condition has become more prevalent as rates of metabolic syndrome, type 2 diabetes, and obesity have also escalated. The unfortunate outcome for many people is liver cirrhosis that warrants transplantation or being unable to receive a transplant since many livers are discarded due to high levels of steatosis. Over the past several years, however, a great deal of work has gone into understanding the pathophysiology of this disease as well as possible treatment options. This review summarizes various defatting strategies including in vitro use of pharmacologic agents, machine perfusion of extracted livers, and genomic approaches targeting specific proteins. The goal of the field is to reduce the number of necessary transplants and expand the pool of organs available for use.
Collapse
|
10
|
Upchurch CM, Yeudall S, Pavelec CM, Merk D, Greulich J, Manjegowda M, Raghavan SS, Bochkis IM, Scott MM, Perez-Reyes E, Leitinger N. Targeting oxidized phospholipids by AAV-based gene therapy in mice with established hepatic steatosis prevents progression to fibrosis. SCIENCE ADVANCES 2022; 8:eabn0050. [PMID: 35857497 PMCID: PMC9286512 DOI: 10.1126/sciadv.abn0050] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 06/03/2022] [Indexed: 05/06/2023]
Abstract
Oxidized phosphatidylcholines (OxPCs) are implicated in chronic tissue damage. Hyperlipidemic LDL-R--deficient mice transgenic for an OxPC-recognizing IgM fragment (scFv-E06) are protected against nonalcoholic fatty liver disease (NAFLD). To examine the effect of OxPC elimination at different stages of NAFLD progression, we used cre-dependent, adeno-associated virus serotype 8-mediated expression of the single-chain variable fragment of E06 (AAV8-scFv-E06) in hepatocytes of albumin-cre mice. AAV8-induced expression of scFv-E06 at the start of FPC diet protected mice from developing hepatic steatosis. Independently, expression of scFv-E06 in mice with established steatosis prevented the progression to hepatic fibrosis. Mass spectrometry-based oxophospho-lipidomics identified individual OxPC species that were reduced by scFv-E06 expression. In vitro, identified OxPC species dysregulated mitochondrial metabolism and gene expression in hepatocytes and hepatic stellate cells. We demonstrate that individual OxPC species independently affect disease initiation and progression from hepatic steatosis to steatohepatitis, and that AAV-mediated expression of scFv-E06 is an effective therapeutic intervention.
Collapse
Affiliation(s)
- Clint M. Upchurch
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Scott Yeudall
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Caitlin M. Pavelec
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Dennis Merk
- Environmentally-Induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Hospital and Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Jan Greulich
- Environmentally-Induced Cardiovascular Degeneration, Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, University Hospital and Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
- IUF-Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Mohan Manjegowda
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Shyam S. Raghavan
- Department of Pathology, University of Virginia, Charlottesville, VA 22904, USA
| | - Irina M. Bochkis
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Michael M. Scott
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Edward Perez-Reyes
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| |
Collapse
|
11
|
Ali HH, AL-Rawi K, Khalaf Y, Alaaraji S, Aldahham B, Awad M, Al-ani O, Al-ani F, Ali AT. Serum Caveolin-1 Level is Inversely Associated with Serum Vaspin, Visfatin, and HbA1c in Newly Diagnosed Men with Type-2 Diabetes. Rep Biochem Mol Biol 2022; 11:299-309. [PMID: 36164629 PMCID: PMC9455185 DOI: 10.52547/rbmb.11.2.299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 02/20/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND The fluctuation in serum caveolin-1 (Cav-1) concentrations is an important indicator of many diseases. Irrespective of the actual cause, a significant reduction of serum Cav-1 is associated with a significant increase in insulin secretion and hyperinsulinemia. The aim of the current study was to evaluate the relationship between serum Cav-1, serum vaspin and visfatin in newly diagnosed men with T2DM. METHODS Eighty-two newly diagnosed men with T2DM were matched for age and body mass indexes (BMIs) with a similar number of non-diabetic men. Serum Cav-1, vaspin and visfatin were assessed through enzyme-linked immunosorbent assay. Fasting serum glucose (FSG), glycohaemoglobin A1C (HbA1c) were both measured using automated method. In addition, waist-circumferences, waist-hip ratio, systolic (SBP), and diastolic blood pressure (DBP) were also obtained. RESULTS Serum concentration of Cav-1(ng/mL) was significantly low in men newly diagnosed with T2DM, (2.334±0.7627) compared with non-diabetic controls (4.321±1.143), p< 0.0001. In contrast, patients with T2DM exhibited significantly higher serum concentrations of vaspin and visfatin (ng/mL), 142.4±60.53) and 2.99±1.091), than controls, 81.53±39.32) and 1.456±0.654), respectively, p< 0.0001. Expectedly, patients with T2DM have significantly higher FSG, HbA1c, systolic blood pressure (SBP), and diastolic blood pressure (DBP). CONCLUSION There was an inverse significant relationship between Cav-1 and vaspin, visfatin, HbA1c, FSG, and hypertension. This study suggests that serum Cav-1 can be used as a diagnostic marker to predict T2DM in individuals and families under high risk.
Collapse
Affiliation(s)
- Hameed Hussein Ali
- Departemnt of Chemistry, College of Sciences, University of Anbar, Ramadi, Al-Anbar province Iraq.
| | - Khalid AL-Rawi
- Departemnt of Chemistry, College of Sciences, University of Anbar, Ramadi, Al-Anbar province Iraq.
| | - Yousif Khalaf
- Department of Clinical Laboratory Sciences, College of Pharmacy, University of Anbar, Ramadi, Al-Anbar province, Iraq.
| | - Shakir Alaaraji
- Department of Chemistry, College of Education for Pure Sciences, University of Anbar, Ramadi, Al-Anbar province, Iraq.
| | - Bilal Aldahham
- Department of Applied Chemistry, College of Applied Sciences, University of Anbar, Ramadi, Al-Anbar province, Iraq.
| | - Muthanna Awad
- Department of Chemistry, College of Education for Pure Sciences, University of Anbar, Ramadi, Al-Anbar province, Iraq.
| | - Osamah Al-ani
- Odessa National Medical University, Odessa, 65000, Ukraine.
| | - Faisal Al-ani
- Odessa National Medical University, Odessa, 65000, Ukraine.
| | | |
Collapse
|
12
|
Chemerin Overexpression in the Liver Protects against Inflammation in Experimental Non-Alcoholic Steatohepatitis. Biomedicines 2022; 10:biomedicines10010132. [PMID: 35052810 PMCID: PMC8773259 DOI: 10.3390/biomedicines10010132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/03/2022] [Accepted: 01/05/2022] [Indexed: 02/01/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is marked by macrophage infiltration and inflammation. Chemerin is a chemoattractant protein and is abundant in hepatocytes. The aim of this study was to gain insight into the role of hepatocyte-produced prochemerin in NASH. Therefore, mice were infected with adeno-associated virus 8 to direct hepatic overexpression of prochemerin in a methionine–choline deficient dietary model of NASH. At the end of the study, hepatic and serum chemerin were higher in the chemerin-expressing mice. These animals had less hepatic oxidative stress, F4/80 and CC-chemokine ligand 2 (CCL2) protein, and mRNA levels of inflammatory genes than the respective control animals. In order to identify the underlying mechanisms, prochemerin was expressed in hepatocytes and the hepatic stellate cells, LX-2. Here, chemerin had no effect on cell viability, production of inflammatory, or pro-fibrotic factors. Notably, cultivation of human peripheral blood mononuclear cells (PBMCs) in the supernatant of Huh7 cells overexpressing chemerin reduced CCL2, interleukin-6, and osteopontin levels in cell media. CCL2 was also low in RAW264.7 cells exposed to Hepa1–6 cell produced chemerin. In summary, the current study showed that prochemerin overexpression had little effect on hepatocytes and hepatic stellate cells. Of note, hepatocyte-produced chemerin deactivated PBMCs and protected against inflammation in experimental NASH.
Collapse
|
13
|
Chang YW, Sung FC, Tzeng YL, Mou CH, Tien PT, Su CW, Teng YK. Risk of Glaucoma Associated with Components of Metabolic Disease in Taiwan: A Nationwide Population-Based Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 19:305. [PMID: 35010564 PMCID: PMC8744805 DOI: 10.3390/ijerph19010305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/08/2021] [Accepted: 12/23/2021] [Indexed: 06/14/2023]
Abstract
PURPOSE This retrospective cohort study was conducted to determine the glaucoma risk associated with metabolic disease (MetD) using insurance claims data of Taiwan. METHODS From the database, we identified patients with newly diagnosed hypertension, diabetes and/or hyperlipidemia from the years 2000 to 2002 as the MetD cohort (N = 42,036) and an age-gender-diagnosis-date matched control cohort without MetD with a two-fold sample size than that of the MetD cohort. Both cohorts were followed until the development of glaucoma, death, or withdrawal, until 31 December 2013. The incidence of glaucoma, and the Cox method estimated hazard ratio (HR) of glaucoma were calculated. Results showed that the incidence of glaucoma was two-fold higher in the MetD cohort than in the controls (1.99 versus 0.99 per 1000 person-years), with an adjusted HR of 1.66 (95% CI: 1.50-1.85). The glaucoma incidence was higher in patients with diabetes than those with hypertension and hyperlipidemia (2.38 versus 1.95 and 1.72 per 1000 person-years, respectively). The incidence increased to 5.67 per 1000 person-years in patients with all three comorbidities, with an aHR of 4.95 (95% CI: 2.35-10.40). We also found higher incidence rates of primary open-angle glaucoma and primary angle-closure glaucoma with aHRs of 2.03 and 1.44, respectively. It was concluded that glaucoma risk increased with the number of MetD. Health providers need to monitor patients with MetD to prevent glaucoma.
Collapse
Affiliation(s)
- Ya-Wen Chang
- School of Nursing, China Medical University, Taichung 406040, Taiwan; (Y.-W.C.); (Y.-L.T.)
| | - Fung-Chang Sung
- Department of Health Services Administration, China Medical University, Taichung 406040, Taiwan;
- Management Office for Health Data, China Medical University Hospital, Taichung 404332, Taiwan;
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung 41354, Taiwan
| | - Ya-Ling Tzeng
- School of Nursing, China Medical University, Taichung 406040, Taiwan; (Y.-W.C.); (Y.-L.T.)
| | - Chih-Hsin Mou
- Management Office for Health Data, China Medical University Hospital, Taichung 404332, Taiwan;
| | - Peng-Tai Tien
- Department of Ophthalmology, China Medical University Hospital, Taichung 404332, Taiwan;
| | - Cheng-Wen Su
- Department of Ophthalmology, Asia University Hospital, Taichung 41354, Taiwan;
| | - Yu-Kuei Teng
- School of Nursing, China Medical University, Taichung 406040, Taiwan; (Y.-W.C.); (Y.-L.T.)
| |
Collapse
|
14
|
Wang J, Jiang W, Xin J, Xue W, Shi C, Wen J, Huang Y, Hu C. Caveolin-1 Alleviates Acetaminophen-Induced Fat Accumulation in Non-Alcoholic Fatty Liver Disease by Enhancing Hepatic Antioxidant Ability via Activating AMPK Pathway. Front Pharmacol 2021; 12:717276. [PMID: 34305621 PMCID: PMC8293675 DOI: 10.3389/fphar.2021.717276] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 06/21/2021] [Indexed: 12/17/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is an independent risk factor for acute liver injury caused by overuse of acetaminophen (APAP). Caveolin-1 (CAV1), a regulator of hepatic energy metabolism and oxidative stress, was found to have a protective effect against NAFLD in our previous study. However, it remains unclear whether CAV1 has a protective effect against APAP-induced hepatotoxicity in NAFLD. The aim of this study was to determine whether CAV1 inhibits oxidative stress through the AMPK/Nrf2/HO-1 pathway to protect the liver from fat accumulation exacerbated by APAP in NAFLD. In this study, seven-week-old C57BL/6 male mice (18–20 g) were raised under similar conditions for in vivo experiment. In vitro, L02 cells were treated with A/O (alcohol and oleic acid mixture) for 48 h, and APAP was added at 24 h for further incubation. The results showed that the protein expression of the AMPK/Nrf2 pathway was enhanced after CAV1 upregulation. The effects of CAV1 on fat accumulation, ROS, and the AMPK/Nrf2 anti-oxidative pathway were reduced after the application of CAV1-siRNA. Finally, treatment with compound C (an AMPK inhibitor) prevented CAV1 plasmid-mediated alleviation of oxidative stress and fat accumulation and reduced the protein level of Nrf2 in the nucleus, demonstrating that the AMPK/Nrf2/HO-1 pathway was involved in the protective effect of CAV1. These results indicate that CAV1 exerted a protective effect against APAP-aggravated lipid deposition and hepatic injury in NAFLD by inhibiting oxidative stress. Therefore, the upregulation of CAV1 might have clinical benefits in reducing APAP-aggravated hepatotoxicity in NAFLD.
Collapse
Affiliation(s)
- Jiarong Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China.,School of Pharmacy, Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,Anhui Institute of Innovative Drugs, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Wei Jiang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China.,School of Pharmacy, Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,Anhui Institute of Innovative Drugs, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Jiao Xin
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China.,School of Pharmacy, Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,Anhui Institute of Innovative Drugs, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Weiju Xue
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China.,School of Pharmacy, Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,Anhui Institute of Innovative Drugs, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Congjian Shi
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China.,School of Pharmacy, Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,Anhui Institute of Innovative Drugs, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Jiagen Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China.,School of Pharmacy, Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,Anhui Institute of Innovative Drugs, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Yan Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China.,School of Pharmacy, Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,Anhui Institute of Innovative Drugs, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| | - Chengmu Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China.,School of Pharmacy, Institute for Liver Diseases of Anhui Medical University, Anhui Medical University, Hefei, China.,Anhui Institute of Innovative Drugs, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, China
| |
Collapse
|
15
|
How does hepatic lipid accumulation lead to lipotoxicity in non-alcoholic fatty liver disease? Hepatol Int 2021; 15:21-35. [PMID: 33548031 PMCID: PMC7886759 DOI: 10.1007/s12072-020-10121-2] [Citation(s) in RCA: 225] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD), characterized as excess lipid accumulation in the liver which is not due to alcohol use, has emerged as one of the major health problems around the world. The dysregulated lipid metabolism creates a lipotoxic environment which promotes the development of NAFLD, especially the progression from simple steatosis (NAFL) to non-alcoholic steatohepatitis (NASH). PURPOSEAND AIM This review focuses on the mechanisms of lipid accumulation in the liver, with an emphasis on the metabolic fate of free fatty acids (FFAs) in NAFLD and presents an update on the relevant cellular processes/mechanisms that are involved in lipotoxicity. The changes in the levels of various lipid species that result from the imbalance between lipolysis/lipid uptake/lipogenesis and lipid oxidation/secretion can cause organellar dysfunction, e.g. ER stress, mitochondrial dysfunction, lysosomal dysfunction, JNK activation, secretion of extracellular vesicles (EVs) and aggravate (or be exacerbated by) hypoxia which ultimately lead to cell death. The aim of this review is to provide an overview of how abnormal lipid metabolism leads to lipotoxicity and the cellular mechanisms of lipotoxicity in the context of NAFLD.
Collapse
|
16
|
Asimakopoulou A, Engel KM, Gassler N, Bracht T, Sitek B, Buhl EM, Kalampoka S, Pinoé-Schmidt M, van Helden J, Schiller J, Weiskirchen R. Deletion of Perilipin 5 Protects Against Hepatic Injury in Nonalcoholic Fatty Liver Disease via Missing Inflammasome Activation. Cells 2020; 9:1346. [PMID: 32481590 PMCID: PMC7348929 DOI: 10.3390/cells9061346] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 12/21/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a leading cause of chronic liver diseases with an increasing prevalence due to rising rates of obesity, metabolic syndrome and type II diabetes. Untreated NAFLD may progress to steatohepatitis (NASH) and ultimately liver cirrhosis. NAFLD is characterized by lipid accumulation, and when sufficient excess lipids are obtained, irreversible liver injury may follow. Perilipin 5 (PLIN5), a known lipid droplet coating protein and triglyceride metabolism regulator, is highly expressed in oxidatively modified tissues but it is still unclear how it affects NAFLD/NASH progress. We here studied how PLIN5 affects NAFLD development induced by a 30-week high-fat diet (HFD) administration in wild type and PLIN5 knock out (Plin5-/-) mice. The disruption of PLIN5 induced differences in lipid metabolism during HFD feeding and was associated with reduced hepatic fat accumulation. Surprisingly, Plin5-/- mice showed mitigated activation of the NLR family pyrin domain-containing 3 (NLRP3) inflammasome, leading to minor hepatic damage. We conclude that PLIN5 is a pleiotropic regulator of hepatic homeostasis in NASH development. Targeting the PLIN5 expression appears critical for protecting the liver from inflammatory activation during chronic NAFLD.
Collapse
Affiliation(s)
- Anastasia Asimakopoulou
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, 52074 Aachen, Germany; (S.K.); (M.P.-S.)
| | - Kathrin M. Engel
- Institute for Medical Physics and Biophysics, Leipzig University, Medical Faculty, 04107 Leipzig, Germany; (K.M.E.); (J.S.)
| | - Nikolaus Gassler
- Section Pathology, Institute of Legal Medicine, University Hospital Jena, 07747 Jena, Germany;
| | - Thilo Bracht
- Ruhr-University Bochum, Medical Faculty, Medizinisches Proteom-Center, 44801 Bochum, Germany; (T.B.); (B.S.)
- Department of Anesthesia, Intensive Care Medicine, and Pain Therapy, University Hospital Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany
| | - Barbara Sitek
- Ruhr-University Bochum, Medical Faculty, Medizinisches Proteom-Center, 44801 Bochum, Germany; (T.B.); (B.S.)
| | - Eva M. Buhl
- Electron Microscopy Facility, Institute of Pathology, RWTH Aachen University Hospital, 52074 Aachen, Germany;
| | - Stavroula Kalampoka
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, 52074 Aachen, Germany; (S.K.); (M.P.-S.)
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95100 Catania, Italy
| | - Manuela Pinoé-Schmidt
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, 52074 Aachen, Germany; (S.K.); (M.P.-S.)
| | - Josef van Helden
- Laboratory Mönchengladbach—MVZ Dr. Stein and Colleagues, 41169 Mönchengladbach, Germany;
| | - Jürgen Schiller
- Institute for Medical Physics and Biophysics, Leipzig University, Medical Faculty, 04107 Leipzig, Germany; (K.M.E.); (J.S.)
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, 52074 Aachen, Germany; (S.K.); (M.P.-S.)
| |
Collapse
|
17
|
Pol A, Morales-Paytuví F, Bosch M, Parton RG. Non-caveolar caveolins – duties outside the caves. J Cell Sci 2020; 133:133/9/jcs241562. [DOI: 10.1242/jcs.241562] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
ABSTRACT
Caveolae are invaginations of the plasma membrane that are remarkably abundant in adipocytes, endothelial cells and muscle. Caveolae provide cells with resources for mechanoprotection, can undergo fission from the plasma membrane and can regulate a variety of signaling pathways. Caveolins are fundamental components of caveolae, but many cells, such as hepatocytes and many neurons, express caveolins without forming distinguishable caveolae. Thus, the function of caveolins goes beyond their roles as caveolar components. The membrane-organizing and -sculpting capacities of caveolins, in combination with their complex intracellular trafficking, might contribute to these additional roles. Furthermore, non-caveolar caveolins can potentially interact with proteins normally excluded from caveolae. Here, we revisit the non-canonical roles of caveolins in a variety of cellular contexts including liver, brain, lymphocytes, cilia and cancer cells, as well as consider insights from invertebrate systems. Non-caveolar caveolins can determine the intracellular fluxes of active lipids, including cholesterol and sphingolipids. Accordingly, caveolins directly or remotely control a plethora of lipid-dependent processes such as the endocytosis of specific cargoes, sorting and transport in endocytic compartments, or different signaling pathways. Indeed, loss-of-function of non-caveolar caveolins might contribute to the common phenotypes and pathologies of caveolin-deficient cells and animals.
Collapse
Affiliation(s)
- Albert Pol
- Cell Compartments and Signaling Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, 08036, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010, Barcelona, Spain
| | - Frederic Morales-Paytuví
- Cell Compartments and Signaling Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain
| | - Marta Bosch
- Cell Compartments and Signaling Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, 08036, Barcelona, Spain
| | - Robert G. Parton
- Institute for Molecular Bioscience (IMB), The University of Queensland (UQ), Brisbane, Queensland 4072, Australia
- Centre for Microscopy and Microanalysis (CMM) IMB, The University of Queensland (UQ), Brisbane, Queensland 4072, Australia
| |
Collapse
|
18
|
Han M, Nwosu ZC, Piorońska W, Ebert MP, Dooley S, Meyer C. Caveolin-1 Impacts on TGF-β Regulation of Metabolic Gene Signatures in Hepatocytes. Front Physiol 2020; 10:1606. [PMID: 32082178 PMCID: PMC7005071 DOI: 10.3389/fphys.2019.01606] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/23/2019] [Indexed: 12/19/2022] Open
Abstract
Caveolin-1 (CAV1) is a membrane protein associated with metabolism in various cell types. The transforming growth factor beta (TGF-β) is a pro-fibrogenic cytokine in the liver, but its metabolic gene signatures remain unclear to date. We have previously shown that CAV1 alters TGF-β signaling and blocks its pro-apoptotic function. Here, we defined TGF-β-induced metabolic gene signatures in hepatocytes and assessed whether CAV1 abundance affects TGF-β control of those metabolic genes. Microarray analyses of primary hepatocytes after TGF-β stimulation (48 h) showed differential expression of 4224 genes, of which 721 are metabolic genes (adjusted p < 0.001). Functional annotation analysis revealed that TGF-β mainly suppresses metabolic gene network, including genes involved in glutathione, cholesterol, fatty acid, and amino acid metabolism. TGF-β also upregulated several genes related to glycan metabolism and ion transport. In contrast to TGF-β effects, CAV1 knockdown triggered the upregulation of metabolic genes. Immortalized mouse hepatocytes (AML12 cells) were used to validate the gene changes induced by TGF-β stimulation and CAV1 knockdown. Noteworthy, of the TGF-β metabolic target genes, CAV1 modulated the expression of 228 (27%). In conclusion, we present several novel metabolic gene signatures of TGF-β in hepatocytes and show that CAV1 abundance alters almost a third of these genes. These findings could enable a better understanding of TGF-β function in normal and diseased liver especially where differential CAV1 level is implicated.
Collapse
Affiliation(s)
- Mei Han
- Section Molecular Hepatology, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Department of Internal Medicine, The Second Hospital of Dalian Medical University, Dalian, China
| | - Zeribe Chike Nwosu
- Section Molecular Hepatology, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Weronika Piorońska
- Section Molecular Hepatology, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias Philip Ebert
- Section Molecular Hepatology, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Steven Dooley
- Section Molecular Hepatology, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christoph Meyer
- Section Molecular Hepatology, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
19
|
Hepatocyte caveolin-1 modulates metabolic gene profiles and functions in non-alcoholic fatty liver disease. Cell Death Dis 2020; 11:104. [PMID: 32029710 PMCID: PMC7005160 DOI: 10.1038/s41419-020-2295-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 11/08/2022]
Abstract
Caveolin-1 (CAV1) is a crucial regulator of lipid accumulation and metabolism. Previous studies have shown that global Cav1 deficiency affects lipid metabolism and hepatic steatosis. We aimed to analyze the consequences of hepatocyte-specific Cav1 knockout under healthy conditions and upon non-alcoholic fatty liver disease (NAFLD) development. Male and female hepatocyte-specific Cav1 knockout (HepCAV1ko) mice were fed a methionine/choline (MCD) deficient diet for 4 weeks. MCD feeding caused severe hepatic steatosis and slight fibrosis. In addition, liver function parameters, i.e., ALT, AST, and GLDH, were elevated, while cholesterol and glucose level were reduced upon MCD feeding. These differences were not affected by hepatocyte-specific Cav1 knockout. Microarray analysis showed strong differences in gene expression profiles of livers from HepCAV1ko mice compared those of global Cav1 knockout animals. Pathway enrichment analysis identified that metabolic alterations were sex-dimorphically regulated by hepatocyte-specific CAV1. In male HepCAV1ko mice, metabolic pathways were suppressed in NAFLD, whereas in female knockout mice induced. Moreover, gender-specific transcription profiles were modulated in healthy animals. In conclusion, our results demonstrate that hepatocyte-specific Cav1 knockout significantly altered gene profiles, did not affect liver steatosis and fibrosis in NAFLD and that gender had severe impact on gene expression patterns in healthy and diseased hepatocyte-specific Cav1 knockout mice.
Collapse
|
20
|
de Aguiar Greca SC, Kyrou I, Pink R, Randeva H, Grammatopoulos D, Silva E, Karteris E. Involvement of the Endocrine-Disrupting Chemical Bisphenol A (BPA) in Human Placentation. J Clin Med 2020; 9:jcm9020405. [PMID: 32028606 PMCID: PMC7074564 DOI: 10.3390/jcm9020405] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/16/2022] Open
Abstract
Background: Endocrine-disrupting chemicals (EDCs) are environmental chemicals/toxicants that humans are exposed to, interfering with the action of multiple hormones. Bisphenol A (BPA) is classified as an EDC with xenoestrogenic activity with potentially adverse effects in reproduction. Currently, a significant knowledge gap remains regarding the complete spectrum of BPA-induced effects on the human placenta. As such, the present study examined the effects of physiologically relevant doses of BPA in vitro. Methods: qRT-PCR, Western blotting, immunofluorescence, ELISA, microarray analyses, and bioinformatics have been employed to study the effects of BPA using nonsyncytialised (non-ST) and syncytialised (ST) BeWo cells. Results: Treatment with 3 nM BPA led to an increase in cell number and altered the phosphorylation status of p38, an effect mediated primarily via the membrane-bound estrogen receptor (GPR30). Nonbiased microarray analysis identified 1195 and 477 genes that were differentially regulated in non-ST BeWo cells, whereas in ST BeWo cells, 309 and 158 genes had altered expression when treated with 3 and 10 nM, respectively. Enriched pathway analyses in non-ST BeWo identified a leptin and insulin overlap (3 nM), methylation pathways (10 nM), and differentiation of white and brown adipocytes (common). In the ST model, most significantly enriched were the nuclear factor erythroid 2-related factor 2 (NRF2) pathway (3 nM) and mir-124 predicted interactions with cell cycle and differentiation (10 nM). Conclusion: Collectively, our data offer a new insight regarding BPA effects at the placental level, and provide a potential link with metabolic changes that can have an impact on the developing fetus.
Collapse
Affiliation(s)
| | - Ioannis Kyrou
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham B4 7ET, UK;
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Institute of Precision Diagnostics and Translational Medicine, UHCW NHS Trust, Coventry CV4 7AL, UK; (H.R.); (D.G.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Ryan Pink
- Dept of Bio. & Med. Sci., Oxford Brookes University, Oxford OX3 0BP, UK;
| | - Harpal Randeva
- Institute of Precision Diagnostics and Translational Medicine, UHCW NHS Trust, Coventry CV4 7AL, UK; (H.R.); (D.G.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Dimitris Grammatopoulos
- Institute of Precision Diagnostics and Translational Medicine, UHCW NHS Trust, Coventry CV4 7AL, UK; (H.R.); (D.G.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Elisabete Silva
- College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK;
- Correspondence: (E.S.); (E.K.)
| | - Emmanouil Karteris
- College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK;
- Correspondence: (E.S.); (E.K.)
| |
Collapse
|
21
|
Abstract
AbstractDietary protein insufficiency has been linked to excessive TAG storage and non-alcoholic fatty liver disease (NAFLD) in developing countries. Hepatic TAG accumulation following a low-protein diet may be due to altered peroxisomal, mitochondrial and gut microbiota function. Hepatic peroxisomes and mitochondria normally mediate metabolism of nutrients to provide energy and substrates for lipogenesis. Peroxisome biogenesis and activities can be modulated by odd-chain fatty acids (OCFA) and SCFA that are derived from gut bacteria, for example, propionate and butyrate. Also produced during amino acid metabolism by peroxisomes and mitochondria, propionate and butyrate concentrations correlate inversely with risk of obesity, insulin resistance and NAFLD. In this horizon-scanning review, we have compiled available evidence on the effects of protein malnutrition on OCFA production, arising from loss in mitochondrial, peroxisomal and gut microbiota function, and its association with lipid accumulation in the liver. The methyl donor amino acid composition of dietary protein is an important contributor to liver function and lipid storage; the presence and abundance of dietary branched-chain amino acids can modulate the composition and metabolic activity of the gut microbiome and, on the other hand, can affect protective OCFA and SCFA production in the liver. In preclinical animal models fed with low-protein diets, specific amino acid supplementation can ameliorate fatty liver disease. The association between low dietary protein intake and fatty liver disease is underexplored and merits further investigation, particularly in vulnerable groups with dietary protein restriction in developing countries.
Collapse
|
22
|
Dewidar B, Meyer C, Dooley S, Meindl-Beinker N. TGF-β in Hepatic Stellate Cell Activation and Liver Fibrogenesis-Updated 2019. Cells 2019; 8:cells8111419. [PMID: 31718044 PMCID: PMC6912224 DOI: 10.3390/cells8111419] [Citation(s) in RCA: 525] [Impact Index Per Article: 87.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/08/2019] [Accepted: 11/09/2019] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis is an advanced liver disease condition, which could progress to cirrhosis and hepatocellular carcinoma. To date, there is no direct approved antifibrotic therapy, and current treatment is mainly the removal of the causative factor. Transforming growth factor (TGF)-β is a master profibrogenic cytokine and a promising target to treat fibrosis. However, TGF-β has broad biological functions and its inhibition induces non-desirable side effects, which override therapeutic benefits. Therefore, understanding the pleiotropic effects of TGF-β and its upstream and downstream regulatory mechanisms will help to design better TGF-β based therapeutics. Here, we summarize recent discoveries and milestones on the TGF-β signaling pathway related to liver fibrosis and hepatic stellate cell (HSC) activation, emphasizing research of the last five years. This comprises impact of TGF-β on liver fibrogenesis related biological processes, such as senescence, metabolism, reactive oxygen species generation, epigenetics, circadian rhythm, epithelial mesenchymal transition, and endothelial-mesenchymal transition. We also describe the influence of the microenvironment on the response of HSC to TGF-β. Finally, we discuss new approaches to target the TGF-β pathway, name current clinical trials, and explain promises and drawbacks that deserve to be adequately addressed.
Collapse
Affiliation(s)
- Bedair Dewidar
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (B.D.); (C.M.); (S.D.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, 31527 Tanta, Egypt
| | - Christoph Meyer
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (B.D.); (C.M.); (S.D.)
| | - Steven Dooley
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (B.D.); (C.M.); (S.D.)
| | - Nadja Meindl-Beinker
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (B.D.); (C.M.); (S.D.)
- Correspondence: ; Tel.: +49-621-383-4983; Fax: +49-621-383-1467
| |
Collapse
|
23
|
Wang S, Bao X. Hyperlipidemia, Blood Lipid Level, and the Risk of Glaucoma: A Meta-Analysis. Invest Ophthalmol Vis Sci 2019; 60:1028-1043. [PMID: 30897618 DOI: 10.1167/iovs.18-25845] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Previous studies reported that hyperlipidemia and blood lipid levels were associated with glaucoma, ocular hypertension (OHT), and intraocular pressure (IOP). However, studies aimed at investigating this association have yielded conflicting results. Therefore, to shed light on these inconclusive findings, we performed multiple distinct meta-analyses to clarify the association of hyperlipidemia and blood lipid levels with glaucoma, OHT, and IOP. Methods A systematic literature search from Embase, Web of Science, and PubMed was performed to identify relevant studies. To assess the association between hyperlipidemia and glaucoma, we used the pooled odds ratio (OR) with 95% confidence interval (CI). When we assessed the association between blood lipid levels and IOP levels, the pooled mean difference in IOP associated with a 10 mg/dL increase in the blood lipid level was estimated. The pooled difference in IOP was also estimated between patients with and without hyperlipidemia. All the papers that assessed the correlation between hyperlipidemia and glaucoma, between blood lipid levels and IOP levels, and between hyperlipidemia and IOP were included in this meta-analysis. Results We detected a marked association between hyperlipidemia and glaucoma (OR = 1.37; 95% CI = 1.16-1.61), with significant heterogeneity among studies. However, hyperlipidemia was not significantly associated with glaucoma in our analysis of only cross-sectional studies, studies that reported only on hypercholesterolemia patients, studies that were conducted only in North America and Europe, or studies in which normal-tension glaucoma (NTG) patients were included only in the subgroup analyses. The pooled results showed that an increase of 10 mg/dL in blood triglyceride levels would increase the IOP by 0.016 mm Hg (95% CI = 0.009-0.024), with evident heterogeneity between studies (P < 0.001; I2 = 92.0%). The pooled results showed that the blood total cholesterol and low-density lipoprotein-cholesterol (LDL-c) level both had a significant association with IOP. When compared to the patients with nonhyperlipidemia, those with hyperlipidemia had a significantly higher IOP of 0.51 mm Hg (95% CI = 0.18-0.83) (P = 0.001 for heterogeneity; I2 = 81.6%). Conclusions The evidence suggests that hyperlipidemia is significantly associated with an increased risk of glaucoma and that hyperlipidemia and the increased blood lipid levels are associated with increased IOP.
Collapse
Affiliation(s)
- Shiming Wang
- Aier Eye Hospital Group, Ningbo Aier Guangming Eye Hospital, Ningbo, China
| | - Xianyi Bao
- Aier Eye Hospital Group, Wuhan Aier Eye Hospital, Wuhan, China
| |
Collapse
|
24
|
Ipsen DH, Lykkesfeldt J, Tveden-Nyborg P. Molecular mechanisms of hepatic lipid accumulation in non-alcoholic fatty liver disease. Cell Mol Life Sci 2018; 75:3313-3327. [PMID: 29936596 PMCID: PMC6105174 DOI: 10.1007/s00018-018-2860-6] [Citation(s) in RCA: 949] [Impact Index Per Article: 135.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/18/2018] [Accepted: 06/20/2018] [Indexed: 12/17/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is currently the world's most common liver disease, estimated to affect up to one-fourth of the population. Hallmarked by hepatic steatosis, NAFLD is associated with a multitude of detrimental effects and increased mortality. This narrative review investigates the molecular mechanisms of hepatic steatosis in NAFLD, focusing on the four major pathways contributing to lipid homeostasis in the liver. Hepatic steatosis is a consequence of lipid acquisition exceeding lipid disposal, i.e., the uptake of fatty acids and de novo lipogenesis surpassing fatty acid oxidation and export. In NAFLD, hepatic uptake and de novo lipogenesis are increased, while a compensatory enhancement of fatty acid oxidation is insufficient in normalizing lipid levels and may even promote cellular damage and disease progression by inducing oxidative stress, especially with compromised mitochondrial function and increased oxidation in peroxisomes and cytochromes. While lipid export initially increases, it plateaus and may even decrease with disease progression, sustaining the accumulation of lipids. Fueled by lipo-apoptosis, hepatic steatosis leads to systemic metabolic disarray that adversely affects multiple organs, placing abnormal lipid metabolism associated with NAFLD in close relation to many of the current life-style-related diseases.
Collapse
Affiliation(s)
- David Højland Ipsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg C, Denmark
| | - Jens Lykkesfeldt
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg C, Denmark
| | - Pernille Tveden-Nyborg
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg C, Denmark.
| |
Collapse
|
25
|
Chen Z, Nie SD, Qu ML, Zhou D, Wu LY, Shi XJ, Ma LR, Li X, Zhou SL, Wang S, Wu J. The autophagic degradation of Cav-1 contributes to PA-induced apoptosis and inflammation of astrocytes. Cell Death Dis 2018; 9:771. [PMID: 29991726 PMCID: PMC6039485 DOI: 10.1038/s41419-018-0795-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 06/04/2018] [Accepted: 06/06/2018] [Indexed: 12/23/2022]
Abstract
The accumulation of palmitic acid (PA), implicated in obesity, can induce apoptotic cell death and inflammation of astrocytes. Caveolin-1 (Cav-1), an essential protein for astrocytes survival, can be degraded by autophagy, which is a double-edge sword that can either promote cell survival or cell death. The aim of this study was to delineate whether the autophagic degradation of Cav-1 is involved in PA-induced apoptosis and inflammation in hippocampal astrocytes. In this study we found that: (1) PA caused apoptotic death and inflammation by autophagic induction; (2) Cav-1 was degraded by PA-induced autophagy and PA induced autophagy in a Cav-1-independent manner; (3) the degradation of Cav-1 was responsible for PA-induced autophagy-dependent apoptotic cell death and inflammation; (4) chronic high-fat diet (HFD) induced Cav-1 degradation, apoptosis, autophagy, and inflammation in the hippocampal astrocytes of rats. Our results suggest that the autophagic degradation of Cav-1 contributes to PA-induced apoptosis and inflammation of astrocytes. Therefore, Cav-1 may be a potential therapeutic target for central nervous system injuries caused by PA accumulation.
Collapse
Affiliation(s)
- Zi Chen
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Sheng-Dan Nie
- Institute of Clinical Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Min-Li Qu
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Di Zhou
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China
| | - Liang-Yan Wu
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Xia-Jie Shi
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Ling-Ran Ma
- Institute of Clinical Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Xin Li
- Institute of Clinical Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Shan-Lei Zhou
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Shan Wang
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China.
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China.
| | - Jing Wu
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China.
| |
Collapse
|