1
|
Wu SA, Li ZJ, Qi L. Endoplasmic reticulum (ER) protein degradation by ER-associated degradation and ER-phagy. Trends Cell Biol 2025:S0962-8924(25)00002-9. [PMID: 39909774 DOI: 10.1016/j.tcb.2025.01.002] [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: 08/09/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 02/07/2025]
Abstract
Protein misfolding and aggregation in the endoplasmic reticulum (ER) have been causally linked to a variety of human diseases. Two key pathways for eliminating misfolded proteins and aggregates in the ER are ER-associated degradation (ERAD) and ER-phagy, respectively. While both pathways have been well characterized biochemically, our understanding of their physiological relevance and significance remains limited. In recent years, significant advances have been made, including the generation and characterization of various knockout and knockin mouse models, the identification of human disease-associated or -causing variants, and insights into the coordination between ERAD and autophagy in physiological contexts. In this review, we summarize these advancements, highlighting the key roles of a highly conserved suppressor of lin-12-like-hydroxymethyl glutaryl-coenzyme A reductase degradation 1 (SEL1L-HRD1) protein complex of ERAD and ER-phagy in health and disease.
Collapse
Affiliation(s)
- Shuangcheng Alivia Wu
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA 22903, USA.
| | - Zexin Jason Li
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA 22903, USA; Medical Scientist Training Program, University of Virginia, School of Medicine, Charlottesville, VA 22903, USA.
| | - Ling Qi
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA 22903, USA.
| |
Collapse
|
2
|
Su B, Fan Z, Wu J, Zhan H. Genetic association of lipid-lowering drug target genes with pancreatic cancer: a Mendelian randomization study. Sci Rep 2025; 15:3282. [PMID: 39863728 PMCID: PMC11762976 DOI: 10.1038/s41598-025-87490-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
Previous studies have found that dyslipidemia is a risk factor for pancreatic cancer (PC), and that lipid-lowering drugs may reduce the risk of PC. However, it is not clear whether dyslipidemia causes PC. The Mendelian randomization (MR) study aimed to investigate the causal role of lipid traits in pancreatic cancer and to assess the potential impact of lipid-lowering drug targets on pancreatic cancer. Genetic variants associated with lipid traits and variants of genes encoding lipid-lowering drug targets were extracted from the Global Lipids Genetics Consortium genome-wide association study (GWAS). Summary statistics for PC were obtained from an independent GWAS datasets. Colocalization analyses were performed to validate the robustness of the results. No significant effect of lipid-lowering drug targets on PC risk was found. Genetic mimicry of lipoprotein lipase (LPL) was potentially associated with PC risks. Significant MR associations were observed in the discovery dataset (OR 1.64 [95% CI 1.24-2.16], p = 4.48*10-4) with PC in one dataset. However, the finding was not verified in the replication dataset. Our findings do not support dyslipidemia as a causal factor for PC. Among lipid-lowering drug targets, LPL is the potential drug target in PC.
Collapse
Affiliation(s)
- Bohan Su
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Zhiyao Fan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Jiexi Wu
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Hanxiang Zhan
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, China.
| |
Collapse
|
3
|
Silbernagel G, Chen YQ, Li H, Lemen D, Wen Y, Zhen EY, Rief M, Kleber ME, Delgado GE, Sarzynski MA, Qian YW, Schmidt B, Erbel R, Trampisch US, Moissl AP, Rudolf H, Schunkert H, Stang A, März W, Trampisch HJ, Scharnagl H, Konrad RJ. Associations of Circulating ANGPTL3, C-Terminal Domain-Containing ANGPTL4, and ANGPTL3/8 and ANGPTL4/8 Complexes with LPL Activity, Diabetes, Inflammation, and Cardiovascular Mortality. Circulation 2025; 151:218-234. [PMID: 39392008 DOI: 10.1161/circulationaha.124.069272] [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: 02/29/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND ANGPTL3/4/8 (angiopoietin-like proteins 3, 4, and 8) are important regulators of LPL (lipoprotein lipase). ANGPTL8 forms complexes with ANGPTL3 and ANGPTL4. ANGPTL4/8 complex formation converts ANGPTL4 from a furin substrate to a plasmin substrate, and both cleavages generate similar C-terminal domain-containing (CD)-ANGPTL4 fragments. Whereas several studies have investigated associations of free ANGPTL proteins with cardiovascular risk, there are no data describing associations of the complexes and CD-ANGPTL4 with outcomes or describing the effects of the complexes on LPL bound to GPIHBP1 (glycosylphosphatidylinositol HDL-binding protein 1). METHODS Recombinant protein assays were used to study ANGPTL protein and complex effects on GPIHBP1-LPL activity. ANGPTL3/8, ANGPTL3, ANGPTL4/8, and CD-ANGPTL4 were measured with dedicated immunoassays in 2394 LURIC (Ludwigshafen Risk and Cardiovascular Health) study participants undergoing coronary angiography and 6188 getABI study (German Epidemiological Trial on Ankle Brachial Index) participants undergoing ankle brachial index measurement. There was a follow-up for cardiovascular death with a median (interquartile range) duration of 9.80 (8.75-10.40) years in the LURIC study and 7.06 (7.00-7.14) years in the getABI study. RESULTS ANGPTL3/8 potently inhibited GPIHBP1-LPL activity and showed positive associations with LDL-C (low-density lipoprotein cholesterol) and triglycerides (both P<0.001). However, in neither study did ANGPTL3/8 correlate with cardiovascular death. Free ANGPTL3 was positively associated with cardiovascular death in the getABI study but not the LURIC study. ANGPTL4/8 and especially CD-ANGPTL4 were positively associated with the prevalence of diabetes, CRP (C-reactive protein; all P<0.001), and cardiovascular death in both studies. In the LURIC and getABI studies, respective hazard ratios for cardiovascular mortality comparing the third with the first ANGPTL4/8 tertile were 1.47 (1.15-1.88) and 1.68 (1.25-2.27) when adjusted for sex, age, body mass index, and diabetes. For CD-ANGPTL4, these hazard ratios were 2.44 (1.86-3.20) and 2.76 (2.00-3.82). CONCLUSIONS ANGPTL3/8 potently inhibited GPIHBP1-LPL enzymatic activity, consistent with its positive association with serum lipids. However, ANGPTL3/8, LDL-C, and triglyceride levels were not associated with cardiovascular death in the LURIC and getABI cohorts. In contrast, concentrations of ANGPTL4/8 and particularly CD-ANGPTL4 were positively associated with inflammation, the prevalence of diabetes, and cardiovascular mortality.
Collapse
Affiliation(s)
- Günther Silbernagel
- Division of Angiology, Department of Internal Medicine (G.S.), Medical University of Graz, Austria
| | - Yan Q Chen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (Y.Q.C., H.L., D.L., Y.W., E.Y.Z., Y.Q., R.J.K.)
| | - Hongxia Li
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (Y.Q.C., H.L., D.L., Y.W., E.Y.Z., Y.Q., R.J.K.)
| | - Deven Lemen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (Y.Q.C., H.L., D.L., Y.W., E.Y.Z., Y.Q., R.J.K.)
| | - Yi Wen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (Y.Q.C., H.L., D.L., Y.W., E.Y.Z., Y.Q., R.J.K.)
| | - Eugene Y Zhen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (Y.Q.C., H.L., D.L., Y.W., E.Y.Z., Y.Q., R.J.K.)
| | - Martin Rief
- Anaesthesiology and Intensive Care Medicine (M.R.), Medical University of Graz, Austria
| | - Marcus E Kleber
- 5th Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (W.M., M.E.K., G.E.D.)
- Synlab Medizinisches Versorgungszentrum Humangenetik Mannheim, GmbH, Germany (M.E.K.)
| | - Graciela E Delgado
- 5th Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (W.M., M.E.K., G.E.D.)
| | - Mark A Sarzynski
- Department of Exercise Science, University of South Carolina, Columbia (M.A.S.)
| | - Yue-Wei Qian
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (Y.Q.C., H.L., D.L., Y.W., E.Y.Z., Y.Q., R.J.K.)
| | - Boerge Schmidt
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University Duisburg-Essen, Germany (A.S., B.S., R.E.)
| | - Raimund Erbel
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University Duisburg-Essen, Germany (A.S., B.S., R.E.)
| | - Ulrike S Trampisch
- Department of Medical Informatics, Biometry and Epidemiology, Ruhr University, Bochum, Germany (U.S.T., H.J.T.)
| | - Angela P Moissl
- Institute of Nutritional Sciences Friedrich Schiller University and Competence Cluster for Nutrition and Cardiovascular Health, Halle-Jena-Leipzig, Jena, Germany (A.P.M.)
| | - Henrik Rudolf
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University, Germany (H.R.)
| | - Heribert Schunkert
- Department of Cardiology, German Heart Center Munich, Technical University of Munich and Partner Site Munich Heart Alliance, German Center for Cardiovascular Disease, Germany (H.S.)
| | - Andreas Stang
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University Duisburg-Essen, Germany (A.S., B.S., R.E.)
- School of Public Health, Department of Epidemiology, Boston University, MA (A.S.)
| | - Winfried März
- Clinical Institute of Medical and Chemical Laboratory Diagnostics (W.M., H.S.), Medical University of Graz, Austria
- 5th Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany (W.M., M.E.K., G.E.D.)
- Synlab Academy, Synlab Holding Germany, Mannheim (W.M.)
| | - Hans J Trampisch
- Department of Medical Informatics, Biometry and Epidemiology, Ruhr University, Bochum, Germany (U.S.T., H.J.T.)
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics (W.M., H.S.), Medical University of Graz, Austria
| | - Robert J Konrad
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (Y.Q.C., H.L., D.L., Y.W., E.Y.Z., Y.Q., R.J.K.)
| |
Collapse
|
4
|
Zhou M, Lv J, Chen X, Shi Y, Chao G, Zhang S. From gut to liver: Exploring the crosstalk between gut-liver axis and oxidative stress in metabolic dysfunction-associated steatotic liver disease. Ann Hepatol 2025; 30:101777. [PMID: 39832564 DOI: 10.1016/j.aohep.2025.101777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/05/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD), now recognized as metabolic dysfunction-associated steatotic liver disease (MASLD), represents a significant and escalating global health challenge. Its prevalence is intricately linked to obesity, insulin resistance, and other components of the metabolic syndrome. As our comprehension of MASLD deepens, it has become evident that this condition extends beyond the liver, embodying a complex, multi-systemic disease with hepatic manifestations that mirror the broader metabolic landscape. This comprehensive review delves into the critical interplay between the gut-liver axis and oxidative stress, elucidating their pivotal roles in the etiology and progression of MASLD. Our analysis reveals several key findings: (1) Bile acid dysregulation can trigger oxidative stress through enhanced ROS production in hepatocytes and Kupffer cells, leading to mitochondrial dysfunction and lipid peroxidation; (2) Gut microbiota dysbiosis disrupts intestinal barrier function, allowing increased translocation of endotoxins like LPS, which activate inflammatory pathways through TLR4 signaling and promote oxidative stress via NADPH oxidase activation; (3) The redox-sensitive transcription factors NF-κB and Nrf2 serve as crucial mediators in the gut-liver axis, with NF-κB regulating inflammatory responses and Nrf2 orchestrating antioxidant defenses; (4) Oxidative stress-induced damage to intestinal barrier function creates a destructive feedback loop, further exacerbating liver inflammation and disease progression. These findings highlight the complex interrelationship between gut-liver axis dysfunction and oxidative stress in MASLD pathogenesis, suggesting potential therapeutic targets for disease management.
Collapse
Affiliation(s)
- Mi Zhou
- Department of Gastroenterology, Xinhua Hospital of zhejiang Province: The Second Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China
| | - Jianyu Lv
- Department of Gastroenterology, Xinhua Hospital of zhejiang Province: The Second Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China
| | - Xinli Chen
- Department of Gastroenterology, Xinhua Hospital of zhejiang Province: The Second Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China
| | - Yujie Shi
- Department of Gastroenterology, Xinhua Hospital of zhejiang Province: The Second Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China
| | - Guanqun Chao
- Department of General Practice, Zhejiang University School of Medicine Sir Run Shaw Hospital, China
| | - Shuo Zhang
- Department of Gastroenterology, Xinhua Hospital of zhejiang Province: The Second Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, China.
| |
Collapse
|
5
|
Li X, Tian Z, Li H, Tan L, Zhang Y, Ge C, Wang K. Impact of Ultraviolet Radiation on Skin and Blood Melanin Traits in Xichou Black-Boned Chicken: A Transcriptomic and Metabolomic Study. Animals (Basel) 2025; 15:141. [PMID: 39858141 PMCID: PMC11758654 DOI: 10.3390/ani15020141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
This study investigated the effects of ultraviolet A (UVA) radiation on the blackness traits of Xichou Black-boned Chickens and their underlying molecular mechanisms through combined transcriptomic and metabolomic analyses. A total of 240 one-day-old Xichou Black-boned Chickens were randomly divided into four groups and exposed to different durations of UVA radiation at an intensity of 47 μW/cm2. Skin blackness was measured at 1, 22, and 45 days of age, and blood and pectoral skin samples were collected at 45 days for analysis. The results showed that an exposure time of 1 h per day at 47 μW/cm2 was ideal for enhancing the blackness traits of the chickens, while exposure times exceeding 3 h led to a significant increase in mortality. Omics studies revealed that the mTOR pathway and tryptophan metabolism pathway are potentially involved in the biological processes by which UVA radiation affects the blackness traits of Xichou Black-boned Chickens, with the FZD3 gene being a candidate gene for these effects. These findings provide theoretical foundations and references for understanding the molecular regulation of blackness traits in these chickens and the potential application of UVA radiation in their production.
Collapse
Affiliation(s)
| | | | | | | | | | - Changrong Ge
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.L.); (Z.T.); (H.L.); (L.T.); (Y.Z.)
| | - Kun Wang
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (X.L.); (Z.T.); (H.L.); (L.T.); (Y.Z.)
| |
Collapse
|
6
|
Perera SD, Wang J, McIntyre AD, Hegele RA. Lipoprotein Lipase: Structure, Function, and Genetic Variation. Genes (Basel) 2025; 16:55. [PMID: 39858602 PMCID: PMC11764694 DOI: 10.3390/genes16010055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 12/28/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
Biallelic rare pathogenic loss-of-function (LOF) variants in lipoprotein lipase (LPL) cause familial chylomicronemia syndrome (FCS). Heterozygosity for these same variants is associated with a highly variable plasma triglyceride (TG) phenotype ranging from normal to severe hypertriglyceridemia (HTG), with longitudinal variation in phenotype severity seen often in a given carrier. Here, we provide an updated overview of genetic variation in LPL in the context of HTG, with a focus on disease-causing and/or disease-associated variants. We provide a curated list of 300 disease-causing variants discovered in LPL, as well as an exon-by-exon breakdown of the LPL gene and protein, highlighting the impact of variants and the various functional residues of domains of the LPL protein. We also provide a curated list of variants of unknown or uncertain significance, many of which may be upgraded to pathogenic/likely pathogenic classification should an additional case and/or segregation data be reported. Finally, we also review the association between benign/likely benign variants in LPL, many of which are common polymorphisms, and the TG phenotype.
Collapse
Affiliation(s)
- Shehan D. Perera
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, 4288A-1151 Richmond Street North, London, ON N6A 5B7, Canada; (S.D.P.); (J.W.); (A.D.M.)
| | - Jian Wang
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, 4288A-1151 Richmond Street North, London, ON N6A 5B7, Canada; (S.D.P.); (J.W.); (A.D.M.)
| | - Adam D. McIntyre
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, 4288A-1151 Richmond Street North, London, ON N6A 5B7, Canada; (S.D.P.); (J.W.); (A.D.M.)
| | - Robert A. Hegele
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, 4288A-1151 Richmond Street North, London, ON N6A 5B7, Canada; (S.D.P.); (J.W.); (A.D.M.)
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, 1151 Richmond Street North, London, ON N6A 5B7, Canada
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, 1151 Richmond Street North, London, ON N6A 5B7, Canada
| |
Collapse
|
7
|
Xu G, Chu M, Shen S, Miao H, Bai Y, Liu X, Liu W, Song P, Wang L, Fu M, Dang E, Shao S, Wang G. Causal effects of circulating lipids and lipid-lowering drugs on the risk of atopic dermatitis: a mendelian randomization study. Arch Dermatol Res 2024; 317:154. [PMID: 39718588 DOI: 10.1007/s00403-024-03635-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/31/2024] [Accepted: 11/26/2024] [Indexed: 12/25/2024]
Abstract
Lipid metabolism disorders are frequently noted in atopic dermatitis (AD) patients, prompting the long-term use of lipid-lowering drugs. However, the causal effects of circulating lipids and different lipid-lowering drugs on the risk of AD are not thoroughly understood. Using publicly available genome-wide association studies (GWAS) summary data from two different cohorts, a series of Mendelian randomization (MR) analyses were conducted to explore the causal effects of genetically proxied circulating lipids and lipid-lowering drugs on the risk of AD. Statistically, the random-effects inverse-variance-weighted (IVW) model was used as main analysis and several methods were conducted for sensitivity analysis to test the robustness of our results. Our findings revealed reduced risks of AD related to genetically proxied subtilisin/kexin type 9 (PCSK9) inhibition and lipoprotein lipase (LPL) agonist, while an increased AD risk associated with Niemann-Pick C1-like 1 (NPC1L1) inhibition. Circulating lipids and other drug targets did not show significant associations with AD risk. These results were replicated in the validation cohort; sensitivity analyses confirmed the robustness. This MR study suggests that, independent of circulating lipids, the use of PCSK9 inhibitors and LPL agonists may be associated with a decreased risk of AD, while inhibition of NPC1L1 is implicated in an increased risk. These findings may help optimize personalized selection of lipid-lowering drugs for AD patients and those at risk of AD.
Collapse
Affiliation(s)
- Guangquan Xu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Mengyang Chu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Shengxian Shen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Haijun Miao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Yaxing Bai
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Xuan Liu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Wanting Liu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Pu Song
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Lei Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Meng Fu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Erle Dang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China
| | - Shuai Shao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China.
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shannxi, China.
| |
Collapse
|
8
|
Zhang Y, Jiang X, Wang W, Lei L, Sheng R, Li S, Luo J, Liu H, Zhang J, Han X, Li Y, Zhang Y, Wang C, Si S, Jin ZG, Xu Y. ASGR1 Deficiency Inhibits Atherosclerosis in Western Diet-Fed ApoE-/- Mice by Regulating Lipoprotein Metabolism and Promoting Cholesterol Efflux. Arterioscler Thromb Vasc Biol 2024; 44:2428-2449. [PMID: 39387120 PMCID: PMC11593992 DOI: 10.1161/atvbaha.124.321076] [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: 04/09/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Atherosclerosis is the most common cause of cardiovascular diseases. Clinical studies indicate that loss-of-function ASGR1 (asialoglycoprotein receptor 1) is significantly associated with lower plasma cholesterol levels and reduces cardiovascular disease risk. However, the effect of ASGR1 on atherosclerosis remains incompletely understood; whether inhibition of ASGR1 causes liver injury remains controversial. Here, we comprehensively investigated the effects and the underlying molecular mechanisms of ASGR1 deficiency and overexpression on atherosclerosis and liver injury in mice. METHODS We engineered Asgr1 knockout mice (Asgr1-/-), Asgr1 and ApoE double-knockout mice (Asgr1-/-ApoE-/-), and ASGR1-overexpressing mice on an ApoE-/- background and then fed them different diets to assess the role of ASGR1 in atherosclerosis and liver injury. RESULTS After being fed a Western diet for 12 weeks, Asgr1-/-ApoE-/- mice exhibited significantly decreased atherosclerotic lesion areas in the aorta and aortic root sections, reduced plasma VLDL (very-low-density lipoprotein) cholesterol and LDL (low-density lipoprotein) cholesterol levels, decreased VLDL production, and increased fecal cholesterol contents. Conversely, ASGR1 overexpression in ApoE-/- mice increased atherosclerotic lesions in the aorta and aortic root sections, augmented plasma VLDL cholesterol and LDL cholesterol levels and VLDL production, and decreased fecal cholesterol contents. Mechanistically, ASGR1 deficiency reduced VLDL production by inhibiting the expression of MTTP (microsomal triglyceride transfer protein) and ANGPTL3 (angiopoietin-like protein 3)/ANGPTL8 (angiopoietin-like protein 8) but increasing LPL (lipoprotein lipase) activity, increased LDL uptake by increasing LDLR (LDL receptor) expression, and promoted cholesterol efflux through increasing expression of LXRα (liver X receptor-α), ABCA1 (ATP-binding cassette subfamily A member 1), ABCG5 (ATP-binding cassette subfamily G member 5), and CYP7A1 (cytochrome P450 family 7 subfamily A member 1). These underlying alterations were confirmed in ASGR1-overexpressing ApoE-/- mice. In addition, ASGR1 deficiency exacerbates liver injury in Western diet-induced Asgr1-/-ApoE-/- mice and high-fat diet-induced but not normal laboratory diet-induced and high-fat and high-cholesterol diet-induced Asgr1-/- mice, while its overexpression mitigates liver injury in Western diet-induced ASGR1-overexpressing ApoE-/- mice. CONCLUSIONS Inhibition of ASGR1 inhibits atherosclerosis in Western diet-fed ApoE-/- mice, suggesting that inhibiting ASGR1 may serve as a novel therapeutic strategy to treat atherosclerosis and cardiovascular diseases.
Collapse
MESH Headings
- Animals
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Diet, Western
- Disease Models, Animal
- Mice
- Male
- Liver/metabolism
- Liver/pathology
- Cholesterol/blood
- Cholesterol/metabolism
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Asialoglycoprotein Receptor/metabolism
- Asialoglycoprotein Receptor/genetics
- Mice, Knockout
- Aortic Diseases/pathology
- Aortic Diseases/prevention & control
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Plaque, Atherosclerotic
- Aorta/pathology
- Aorta/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 5/metabolism
- Macrophages/metabolism
- Signal Transduction
- Apolipoproteins E/genetics
- Apolipoproteins E/deficiency
Collapse
Affiliation(s)
- Yuyan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC (National Health Commission) Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China (Yuyan Zhang, X.J., W.W., L.L., R.S., S.L., J.L., J.Z., X.H., Y.L., Yuhao Zhang, C.W., S.S., Y.X.)
| | - Xinhai Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC (National Health Commission) Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China (Yuyan Zhang, X.J., W.W., L.L., R.S., S.L., J.L., J.Z., X.H., Y.L., Yuhao Zhang, C.W., S.S., Y.X.)
| | - Weizhi Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC (National Health Commission) Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China (Yuyan Zhang, X.J., W.W., L.L., R.S., S.L., J.L., J.Z., X.H., Y.L., Yuhao Zhang, C.W., S.S., Y.X.)
| | - Lijuan Lei
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC (National Health Commission) Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China (Yuyan Zhang, X.J., W.W., L.L., R.S., S.L., J.L., J.Z., X.H., Y.L., Yuhao Zhang, C.W., S.S., Y.X.)
| | - Ren Sheng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC (National Health Commission) Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China (Yuyan Zhang, X.J., W.W., L.L., R.S., S.L., J.L., J.Z., X.H., Y.L., Yuhao Zhang, C.W., S.S., Y.X.)
| | - Shunwang Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC (National Health Commission) Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China (Yuyan Zhang, X.J., W.W., L.L., R.S., S.L., J.L., J.Z., X.H., Y.L., Yuhao Zhang, C.W., S.S., Y.X.)
| | - Jinque Luo
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC (National Health Commission) Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China (Yuyan Zhang, X.J., W.W., L.L., R.S., S.L., J.L., J.Z., X.H., Y.L., Yuhao Zhang, C.W., S.S., Y.X.)
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.L., H.L., Z.-G.J.)
| | - Huan Liu
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.L., H.L., Z.-G.J.)
| | - Jing Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC (National Health Commission) Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China (Yuyan Zhang, X.J., W.W., L.L., R.S., S.L., J.L., J.Z., X.H., Y.L., Yuhao Zhang, C.W., S.S., Y.X.)
| | - Xiaowan Han
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC (National Health Commission) Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China (Yuyan Zhang, X.J., W.W., L.L., R.S., S.L., J.L., J.Z., X.H., Y.L., Yuhao Zhang, C.W., S.S., Y.X.)
| | - Yining Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC (National Health Commission) Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China (Yuyan Zhang, X.J., W.W., L.L., R.S., S.L., J.L., J.Z., X.H., Y.L., Yuhao Zhang, C.W., S.S., Y.X.)
| | - Yuhao Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC (National Health Commission) Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China (Yuyan Zhang, X.J., W.W., L.L., R.S., S.L., J.L., J.Z., X.H., Y.L., Yuhao Zhang, C.W., S.S., Y.X.)
| | - Chenyin Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC (National Health Commission) Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China (Yuyan Zhang, X.J., W.W., L.L., R.S., S.L., J.L., J.Z., X.H., Y.L., Yuhao Zhang, C.W., S.S., Y.X.)
| | - Shuyi Si
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC (National Health Commission) Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China (Yuyan Zhang, X.J., W.W., L.L., R.S., S.L., J.L., J.Z., X.H., Y.L., Yuhao Zhang, C.W., S.S., Y.X.)
| | - Zheng-Gen Jin
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, NY (J.L., H.L., Z.-G.J.)
| | - Yanni Xu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, NHC (National Health Commission) Key Laboratory of Biotechnology of Antibiotics, National Center for New Microbial Drug Screening, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China (Yuyan Zhang, X.J., W.W., L.L., R.S., S.L., J.L., J.Z., X.H., Y.L., Yuhao Zhang, C.W., S.S., Y.X.)
| |
Collapse
|
9
|
Ou G, Zhang Y, Cai H, Yao K, Qiu Z, Chen Y, Yang Y, Chen Q, Chen X. Lipid-lowering drugs, circulating inflammatory factors, and atrial fibrillation: a mediation Mendelian randomization study. Front Cardiovasc Med 2024; 11:1446610. [PMID: 39563941 PMCID: PMC11573524 DOI: 10.3389/fcvm.2024.1446610] [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: 06/10/2024] [Accepted: 10/14/2024] [Indexed: 11/21/2024] Open
Abstract
Background Previous studies have shown an association between lipid-lowering drugs, circulating inflammatory factors, and atrial fibrillation (AF), but the specific effects of lipid-lowering drugs on AF and whether they can be mediated by circulating inflammatory factors remain unclear. Methods We collected 10 genetic variants encoding lipid-lowering drug targets (LDLR, HMGCR, PCSK9, NPC1L1, APOB, APOB, ABCG5, ABCG8, LPL, APOC3, and PPARA) and AF based on genome-wide association study (GWAS) summary statistics. Drug target Mendelian randomization (MR) was used to explore the causal relationship between lipid-lowering drugs and AF. In addition, we performed a mediation analysis of 91 circulating inflammatory factors to explore potential mediators. Sensitivity analyses were performed to verify the reliability of the MR Results by MR-Egger intercept test, Cochran's Q test and leave-one-out test. Results The results of IVW method showed that LPL agonist had a protective effect on AF(OR = 0. 854, 95%CI: 0.816-0.894, P = 1.844E-11). However, the other nine lipid-lowering drug targets had no significant effect on AF. Notably, we found a mediator role of Fibroblast Growth Factor 5 (FGF5) in the protective effect of LPL agonist on AF with a mediator ratio of 9.22%. Sensitivity analyses supported the robustness of our findings, indicating a possible mediating pathway by which LPL agonists affect the risk of AF. Conclusion Our study provides new insights into the complex interactions among lipid-lowering agents, circulating inflammatory factors and AF, and also identified a potential mediating role of FGF5 in the pathogenesis of AF. Our findings highlight the potential of LPL agonists and targeting specific inflammatory factors for therapeutic intervention in AF, providing promising avenues for future research and clinical strategies for the management and prevention of AF.
Collapse
Affiliation(s)
- Guangyang Ou
- The First Clinical College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yi Zhang
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou Medical University, Jinzhou, China
| | - Huzhi Cai
- International Medical Department, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Kunpeng Yao
- The First Clinical College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Zerui Qiu
- The First Clinical College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yaowu Chen
- The First Clinical College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yang Yang
- The First Clinical College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Qingyang Chen
- International Medical Department, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Xinyu Chen
- Preventive Treatment Center, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
10
|
Chen J, Luo Q, Yi Y, Wang J, Chen P, Luo F, Fang Z. ANGPTL3 as a target for treating lipid disorders in type 2 diabetes patients. Lipids Health Dis 2024; 23:356. [PMID: 39487451 PMCID: PMC11528995 DOI: 10.1186/s12944-024-02352-1] [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: 06/04/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a globally prevalent metabolic disorder, and cardiovascular disease (CVD) is a significant cause of mortality and morbidity in diabetic individuals. In addition to hyperglycemia, lipid abnormalities associated with T2DM play a crucial role in the development of CVD complications. Diabetic dyslipidemia is characterized by elevated levels of triglyceride (TG)-rich lipoproteins and small dense low-density lipoprotein (LDL) particles, reduced high-density lipoprotein (HDL) cholesterol, and impaired HDL function. Angiopoietin protein-like 3 (ANGPTL3) is a liver-derived protein that plays a crucial role in regulating plasma lipoprotein metabolism by inhibiting lipoprotein lipase and influencing lipid levels. Inhibiting ANGPTL3 has shown promising effects in promoting HDL-mediated cholesterol reverse transport and reducing the levels of TG-rich lipoproteins and LDL cholesterol. Here, we explore the potential of ANGPTL3 as a therapeutic target for lipid management in T2DM patients.
Collapse
Affiliation(s)
- Jingfei Chen
- Research Institute of Blood Lipid and Atherosclerosis, Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China
| | - Qin Luo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, 410011, P.R. China
| | - Yanfeng Yi
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, 410011, P.R. China
| | - Jiangang Wang
- Department of Health Management Center, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, P.R. China
| | - Pengfei Chen
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China.
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, 410011, P.R. China.
| | - Fei Luo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China.
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, 410011, P.R. China.
| | - Zhenfei Fang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, P.R. China.
- Research Institute of Blood Lipid and Atherosclerosis, Central South University, Changsha, Hunan, 410011, P.R. China.
| |
Collapse
|
11
|
Miao X, Alidadipour A, Saed V, Sayyadi F, Jadidi Y, Davoudi M, Amraee F, Jadidi N, Afrisham R. Hepatokines: unveiling the molecular and cellular mechanisms connecting hepatic tissue to insulin resistance and inflammation. Acta Diabetol 2024; 61:1339-1361. [PMID: 39031190 DOI: 10.1007/s00592-024-02335-9] [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: 05/04/2024] [Accepted: 07/06/2024] [Indexed: 07/22/2024]
Abstract
Insulin resistance arising from Non-Alcoholic Fatty Liver Disease (NAFLD) stands as a prevalent global ailment, a manifestation within societies stemming from individuals' suboptimal dietary habits and lifestyles. This form of insulin resistance emerges as a pivotal factor in the development of type 2 diabetes mellitus (T2DM). Emerging evidence underscores the significant role of hepatokines, as hepatic-secreted hormone-like entities, in the genesis of insulin resistance and eventual onset of type 2 diabetes. Hepatokines exert influence over extrahepatic metabolism regulation. Their principal functions encompass impacting adipocytes, pancreatic cells, muscles, and the brain, thereby playing a crucial role in shaping body metabolism through signaling to target tissues. This review explores the most important hepatokines, each with distinct influences. Our review shows that Fetuin-A promotes lipid-induced insulin resistance by acting as an endogenous ligand for Toll-like receptor 4 (TLR-4). FGF21 reduces inflammation in diabetes by blocking the nuclear translocation of nuclear factor-κB (NF-κB) in adipocytes and adipose tissue, while also improving glucose metabolism. ANGPTL6 enhances AMPK and insulin signaling in muscle, and suppresses gluconeogenesis. Follistatin can influence insulin resistance and inflammation by interacting with members of the TGF-β family. Adropin show a positive correlation with phosphoenolpyruvate carboxykinase 1 (PCK1), a key regulator of gluconeogenesis. This article delves into hepatokines' impact on NAFLD, inflammation, and T2DM, with a specific focus on insulin resistance. The aim is to comprehend the influence of these recently identified hormones on disease development and their underlying physiological and pathological mechanisms.
Collapse
Affiliation(s)
- Xiaolei Miao
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China
| | - Arian Alidadipour
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Vian Saed
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Firooze Sayyadi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Yasaman Jadidi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Davoudi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Amraee
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Nastaran Jadidi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Afrisham
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Alahmadi H, Martinez S, Farrell R, Bikienga R, Arinzeh N, Potts C, Li Z, Warner GR. Mixtures of phthalates disrupt expression of genes related to lipid metabolism and peroxisome proliferator-activated receptor signaling in mouse granulosa cells. Toxicol Sci 2024; 202:69-84. [PMID: 39150890 PMCID: PMC11514836 DOI: 10.1093/toxsci/kfae105] [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] [Indexed: 08/18/2024] Open
Abstract
Phthalates are a class of known endocrine-disrupting chemicals that are found in common everyday products. Several studies associate phthalate exposure with detrimental effects on ovarian function, including growth and development of the follicle and production of steroid hormones. We hypothesized that dysregulation of the ovary by phthalates may be mediated by phthalate toxicity towards granulosa cells, a major cell type in ovarian follicles responsible for key steps of hormone production and nourishing the developing oocyte. To test the hypothesis that phthalates target granulosa cells, we harvested granulosa cells from adult CD-1 mouse ovaries and cultured them for 96 h in vehicle control, a phthalate mixture, or a phthalate metabolite mixture (0.1 to 100 μg/ml). After culture, we measured metabolism of the phthalate mixture into monoester metabolites by the granulosa cells, finding that granulosa cells do not significantly contribute to ovarian metabolism of phthalates. Immunohistochemistry of phthalate metabolizing enzymes in whole ovaries confirmed that these enzymes are not strongly expressed in granulosa cells of antral follicles and that ovarian metabolism of phthalates likely occurs primarily in the stroma. RNA sequencing of treated granulosa cells identified 407 differentially expressed genes, with overrepresentation of genes from lipid metabolic processes, cholesterol metabolism, and peroxisome proliferator-activated receptor (PPAR) signaling pathways. Expression of significantly differentially expressed genes related to these pathways was confirmed using qPCR. Our results agree with previous findings that phthalates and phthalate metabolites have different effects on the ovary, but both interfere with PPAR signaling in granulosa cells.
Collapse
Affiliation(s)
- Hanin Alahmadi
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, Newark, NJ 07103, United States
| | - Stephanie Martinez
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, Newark, NJ 07103, United States
| | - Rivka Farrell
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, Newark, NJ 07103, United States
| | - Rafiatou Bikienga
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, Newark, NJ 07103, United States
| | - Nneka Arinzeh
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, Newark, NJ 07103, United States
| | - Courtney Potts
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, Newark, NJ 07103, United States
| | - Zhong Li
- Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL 61802, United States
| | - Genoa R Warner
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, Newark, NJ 07103, United States
| |
Collapse
|
13
|
Burks KH, Stitziel NO, Davidson NO. Molecular Regulation and Therapeutic Targeting of VLDL Production in Cardiometabolic Disease. Cell Mol Gastroenterol Hepatol 2024; 19:101409. [PMID: 39406347 PMCID: PMC11609389 DOI: 10.1016/j.jcmgh.2024.101409] [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: 06/20/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 11/16/2024]
Abstract
There exists a complex relationship between steatotic liver disease (SLD) and atherosclerotic cardiovascular disease (CVD). CVD is a leading cause of morbidity and mortality among individuals with SLD, particularly those with metabolic dysfunction-associated SLD (MASLD), a significant proportion of whom also exhibit features of insulin resistance. Recent evidence supports an expanded role of very low-density lipoprotein (VLDL) in the pathogenesis of CVD in patients, both with and without associated metabolic dysfunction. VLDL represents the major vehicle for exporting neutral lipid from hepatocytes, with each particle containing one molecule of apolipoproteinB100 (APOB100). VLDL production becomes dysregulated under conditions characteristic of MASLD including steatosis and insulin resistance. Insulin resistance not only affects VLDL production but also mediates the pathogenesis of atherosclerotic CVD. VLDL assembly and secretion therefore represents an important pathway in the setting of cardiometabolic disease and offers several candidates for therapeutic targeting, particularly in metabolically complex patients with MASLD at increased risk of atherosclerotic CVD. Here we review the clinical significance as well as the translational and therapeutic potential of key regulatory steps impacting VLDL initiation, maturation, secretion, catabolism, and clearance.
Collapse
Affiliation(s)
- Kendall H Burks
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, Saint Louis, Missouri
| | - Nathan O Stitziel
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, Saint Louis, Missouri
| | - Nicholas O Davidson
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri.
| |
Collapse
|
14
|
Kiyamudeen F, Rajapaksha M, Atapattu N, Kularatne SD, Schröder S, Hooper AJ, Burnett JR, Jasinge E. Homozygous LPL and GPIHBP1 variants causing familial chylomicronaemia syndrome in Sri Lankan children. Pathology 2024; 56:904-906. [PMID: 38777740 DOI: 10.1016/j.pathol.2024.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 02/16/2024] [Accepted: 02/28/2024] [Indexed: 05/25/2024]
Affiliation(s)
| | | | - Navoda Atapattu
- Department of Paediatrics, Lady Ridgeway Hospital for Children, Colombo, Sri Lanka
| | | | | | - Amanda J Hooper
- Department of Clinical Biochemistry, PathWest Laboratory Medicine, Royal Perth Hospital and Fiona Stanley Hospital Network, Perth, WA, Australia; School of Medicine, University of Western Australia, Perth, WA, Australia
| | - John R Burnett
- Department of Clinical Biochemistry, PathWest Laboratory Medicine, Royal Perth Hospital and Fiona Stanley Hospital Network, Perth, WA, Australia; School of Medicine, University of Western Australia, Perth, WA, Australia
| | - Eresha Jasinge
- Department of Chemical Pathology, Lady Ridgeway Hospital for Children, Colombo, Sri Lanka
| |
Collapse
|
15
|
Cai W, Pierzynowska K, Stiernborg M, Xu J, Nilsson IA, Svensson U, Melas PA, Lavebratt C. Multispecies synbiotics alleviate dextran sulfate sodium (DSS)-induced colitis: Effects on clinical scores, intestinal pathology, and plasma biomarkers in male and female mice. Clin Nutr ESPEN 2024; 63:74-83. [PMID: 38923468 DOI: 10.1016/j.clnesp.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/04/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is characterized by recurrent inflammation of the gastrointestinal tract and has been linked to an imbalance in gut bacteria. Synbiotics, which combine probiotics and prebiotics, are emerging as potential IBD treatments. AIM To examine the effects of four synbiotic formulations on intestinal inflammation and peripheral biomarkers in a rodent IBD model of both sexes. METHODS Colitis was induced in male and female C57BL/6 mice using 1% dextran sulfate sodium (DSS). Concurrently, a non-exposed control group was maintained. Starting on day 4 post-induction, DSS-exposed mice received one of four synbiotic preparations (Synbio1-4 composed of lactic acid bacteria, Bifidobacterium and dietary fibres), an anti-inflammatory drug used to treat IBD (mesalazine), or placebo (water) until day 14. Clinical symptoms and body weight were monitored daily. Blood samples (taken on days -3, 4, and 14, relative to DSS introduction), were used to analyze plasma biomarkers. At the end of the study, intestinal tissues underwent histological and morphological evaluation. RESULTS Compared to placebo, the Synbio1-, 2- and 3-treated groups had improved clinical scores by day 14. Synbio1 was the only preparation that led to clinical improvements to scores comparable to those of controls. The Synbio1-and 3-treated groups also demonstrated histological improvements in the colon. Plasma biomarker analyses revealed significant Synbio1-induced changes in plasma IL17A, VEGFD, and TNFRSF11B levels that correlated with improved clinical or histological scores. Sex-stratified analyses revealed that most therapeutic-like effects were more pronounced in females. CONCLUSION Our findings underscore the potential therapeutic benefits of specific synbiotics for IBD management. However, further research is needed to validate these outcomes in human subjects.
Collapse
Affiliation(s)
- Wenjie Cai
- Karolinska Institutet, Department of Molecular Medicine and Surgery, Stockholm, Sweden; Karolinska University Hospital Solna, Center for Molecular Medicine, Stockholm, Sweden
| | | | - Miranda Stiernborg
- Karolinska Institutet, Department of Molecular Medicine and Surgery, Stockholm, Sweden; Karolinska University Hospital Solna, Center for Molecular Medicine, Stockholm, Sweden
| | - Jingjing Xu
- Karolinska Institutet, Department of Molecular Medicine and Surgery, Stockholm, Sweden; Karolinska University Hospital Solna, Center for Molecular Medicine, Stockholm, Sweden
| | - Ida Ak Nilsson
- Karolinska Institutet, Department of Molecular Medicine and Surgery, Stockholm, Sweden; Karolinska University Hospital Solna, Center for Molecular Medicine, Stockholm, Sweden
| | | | - Philippe A Melas
- Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, 11364 Stockholm, Sweden
| | - Catharina Lavebratt
- Karolinska Institutet, Department of Molecular Medicine and Surgery, Stockholm, Sweden; Karolinska University Hospital Solna, Center for Molecular Medicine, Stockholm, Sweden.
| |
Collapse
|
16
|
Gugliucci A. Angiopoietin-like Proteins and Lipoprotein Lipase: The Waltz Partners That Govern Triglyceride-Rich Lipoprotein Metabolism? Impact on Atherogenesis, Dietary Interventions, and Emerging Therapies. J Clin Med 2024; 13:5229. [PMID: 39274442 PMCID: PMC11396212 DOI: 10.3390/jcm13175229] [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: 08/22/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Over 50% of patients who take statins are still at risk of developing atherosclerotic cardiovascular disease (ASCVD) and do not achieve their goal LDL-C levels. This residual risk is largely dependent on triglyceride-rich lipoproteins (TRL) and their remnants. In essence, remnant cholesterol-rich chylomicron (CM) and very-low-density lipoprotein (VLDL) particles play a role in atherogenesis. These remnants increase when lipoprotein lipase (LPL) activity is inhibited. ApoCIII has been thoroughly studied as a chief inhibitor and therapeutic options to curb its effect are available. On top of apoCIII regulation of LPL activity, there is a more precise control of LPL in various tissues, which makes it easier to physiologically divide the TRL burden according to the body's requirements. In general, oxidative tissues such as skeletal and cardiac muscle preferentially take up lipids during fasting. Conversely, LPL activity in adipocytes increases significantly after feeding, while its activity in oxidative tissues decreases concurrently. This perspective addresses the recent improvements in our understanding of circadian LPL regulations and their therapeutic implications. Three major tissue-specific lipolysis regulators have been identified: ANGPTL3, ANGPTL4, and ANGPTL8. Briefly, during the postprandial phase, liver ANGPTL8 acts on ANGPTL3 (which is released continuously from the liver) to inhibit LPL in the heart and muscle through an endocrine mechanism. On the other hand, when fasting, ANGPTL4, which is released by adipocytes, inhibits lipoprotein lipase in adipose tissue in a paracrine manner. ANGPTL3 inhibitors may play a therapeutic role in the treatment of hypertriglyceridemia. Several approaches are under development. We look forward to future studies to clarify (a) the nature of hormonal and nutritional factors that determine ANGPTL3, 4, and 8 activities, along with what long-term impacts may be expected if their regulation is impaired pharmacologically; (b) the understanding of the quantitative hierarchy and interaction of the regulatory actions of apoCIII, apoAV, and ANGPTL on LPL activity; (c) strategies for the safe and proper treatment of postprandial lipemia; and (d) the effect of fructose restriction on ANGPTL3, ANGPTL4, and ANGPTL8.
Collapse
Affiliation(s)
- Alejandro Gugliucci
- Glycation, Oxidation and Disease Laboratory, Touro University California, Vallejo, CA 94592, USA
| |
Collapse
|
17
|
Liu MM, Chen X, Bao XH, Huang BH. Lipids, lipid-lowering drugs and lateral epicondylitis of the humerus: a drug-targeted Mendelian randomization study. Front Genet 2024; 15:1437712. [PMID: 39286458 PMCID: PMC11402682 DOI: 10.3389/fgene.2024.1437712] [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: 05/24/2024] [Accepted: 08/22/2024] [Indexed: 09/19/2024] Open
Abstract
Background Clinical observations indicate that blood lipids may be risk factors for lateral epicondylitis (LE) of the humerus, and lipid-lowering drugs are also used for the prevention and treatment of tendon diseases, but these lack high-quality clinical trial evidence and remain inconclusive. Mendelian randomization (MR) analyses can overcome biases in traditional observational studies and offer more accurate inference of causal relationships. Therefore, we employed this approach to investigate whether blood lipids are risk factors for LE and if lipid-lowering drugs can prevent it. Methods Genetic variations associated with lipid traits, including low-density lipoprotein cholesterol (LDL-C), triglycerides (TG), and total cholesterol (TC), were obtained from the UK Biobank and the Global Lipids Genetics Consortium (GLGC). Data on genetic variation in LE were sourced from FinnGen, including 24,061 patients and 275,212 controls. Subsequently, MR analyses were conducted to assess the potential correlation between lipid traits and LE. Additionally, drug-target Mendelian randomization analyses were performed on 10 drug targets relevant to LE. For those drug targets that yielded significant results, further analysis was conducted using colocalization techniques. Results No correlation was found between three blood lipid traits and LE. Lipoprotein lipase (LPL) enhancement is significantly associated with a decreased risk of LE (OR = 0.76, [95% CI, 0.65-0.90], p = 0.001). The expression of LPL in the blood is associated with LE and shares a single causal variant (12.07%), greatly exceeding the probability of different causal variations (1.93%), with a colocalization probability of 86.2%. Conclusion The three lipid traits are not risk factors for lateral epicondylitis. LPL is a potential drug target for the prevention and treatment of LE.
Collapse
Affiliation(s)
- Meng-Meng Liu
- School of Physical Education And Health, Guangxi Medical University, Nanning, China
| | - Xiang Chen
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiao-Hang Bao
- Department of Spinal Surgery, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Bao-Hua Huang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
18
|
Formisano E, Proietti E, Perrone G, Demarco V, Galoppi P, Stefanutti C, Pisciotta L. Characteristics, Physiopathology and Management of Dyslipidemias in Pregnancy: A Narrative Review. Nutrients 2024; 16:2927. [PMID: 39275243 PMCID: PMC11397408 DOI: 10.3390/nu16172927] [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: 07/26/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/16/2024] Open
Abstract
Dyslipidemia is a significant risk factor for atherosclerotic cardiovascular disease (ASCVD). During pregnancy, physiological changes elevate cholesterol and triglyceride levels to support fetal development, which can exacerbate pre-existing conditions and lead to complications such as pre-eclampsia, gestational diabetes, and increased ASCVD risk for both mother and child. Effective management strategies are necessary, especially for pregnant women with inherited forms of dyslipidemia (i.e., familial hypertriglyceridemia, hyperchylomicronemia), where personalized dietary adjustments are crucial for successful pregnancy outcomes. Pharmacological interventions and lipoprotein apheresis may be necessary for severe cases, though their use is often limited by factors such as cost, availability, and potential fetal risks. Despite the promise of advanced therapies, their widespread application remains constrained by limited studies and high costs. Thus, a personalized, multidisciplinary approach is essential for optimizing outcomes. This review provides a comprehensive overview of current strategies and evidence-based practices for managing dyslipidemia during pregnancy, emphasizing the balance of maternal and fetal health. Additionally, it discusses the physiological changes in lipid metabolism during pregnancy and their implications, particularly for women with inherited forms of dyslipidemia.
Collapse
Affiliation(s)
- Elena Formisano
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- Dietetics and Clinical Nutrition Unit, IRCCS Policlinic Hospital San Martino, 16132 Genoa, Italy
| | - Elisa Proietti
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
| | - Giuseppina Perrone
- Department of Maternal and Child Health and Urological Sciences, Umberto I Hospital, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Valentina Demarco
- Department of Maternal and Child Health and Urological Sciences, Umberto I Hospital, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Paola Galoppi
- Department of Maternal and Child Health and Urological Sciences, Umberto I Hospital, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Claudia Stefanutti
- Department of Molecular Medicine, Extracorporeal Therapeutic Techniques Unit, Lipid Clinic and Atherosclerosis Prevention Centre, Regional Centre for Rare Diseases, Immunohematology and Transfusion Medicine, Umberto I Hospital, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Livia Pisciotta
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- Dietetics and Clinical Nutrition Unit, IRCCS Policlinic Hospital San Martino, 16132 Genoa, Italy
| |
Collapse
|
19
|
Tan Y, Huang X, Xu C, Huang Y, Li S, Yin Z. Integrating Genomics and Transcriptomics to Identify Candidate Genes for Egg Production in Taihe Black-Bone Silky Fowls ( Gallus gallus domesticus Brisson). Int J Mol Sci 2024; 25:9373. [PMID: 39273321 PMCID: PMC11395579 DOI: 10.3390/ijms25179373] [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: 07/24/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
The Taihe Black-Bone Silky Fowl (Gallus gallus domesticus Brisson) possesses significant value in terms of consumption, medicinal applications, and ornamental appeal, representing a precious genetic resource and traditional Chinese medicinal material. However, considerable variation exists within populations regarding egg-laying performance. This study integrates a whole-genome selection signal analysis (SSA) with a transcriptome analysis to identify genes associated with egg-laying traits in Taihe Black-Bone Silky Fowls. We identified 31 candidate genes under selection from the high-yield chicken (HC) and low-yield chicken (LC) groups. Additionally, through RNA-seq analysis, 257 common differentially expressed genes (DEGs) were identified from four comparative groups. Two overlapping genes-LPL and SETBP1-were found in both the selected gene and DEG lists. These selected genes and DEGs were enriched in pathways related to ovarian development, including the lysosome pathway, the ECM-receptor interaction pathway, the TGF-beta signaling pathway, the Wnt signaling pathway, the PPAR signaling pathway, and the glycerolipid metabolism pathway. These research findings contribute to the breeding of Taihe Black-Bone Silky Fowls with high egg production traits and provide a theoretical foundation for exploring the regulatory mechanisms of avian reproduction.
Collapse
Affiliation(s)
- Yuting Tan
- College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - Xuan Huang
- College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - Chunhui Xu
- College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - Yunyan Huang
- College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - Shibao Li
- College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - Zhaozheng Yin
- College of Animal Science, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
20
|
Justin Grams R, Yuan K, Founds MW, Ware ML, Pilar MG, Hsu KL. Imidazoles are Tunable Nucleofuges for Developing Tyrosine-Reactive Electrophiles. Chembiochem 2024; 25:e202400382. [PMID: 38819848 PMCID: PMC11462048 DOI: 10.1002/cbic.202400382] [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: 05/08/2024] [Accepted: 05/28/2024] [Indexed: 06/01/2024]
Abstract
Imidazole-1-sulfonyl and -sulfonate (imidazylate) are widely used in synthetic chemistry as nucleofuges for diazotransfer, nucleophilic substitution, and cross-coupling reactions. The utility of these reagents for protein bioconjugation, in contrast, have not been comprehensively explored and important considering the prevalence of imidazoles in biomolecules and drugs. Here, we synthesized a series of alkyne-modified sulfonyl- and sulfonate-imidazole probes to investigate the utility of this electrophile for protein binding. Alkylation of the distal nitrogen activated the nucleofuge capability of the imidazole to produce sulfonyl-imidazolium electrophiles that were highly reactive but unstable for biological applications. In contrast, arylsulfonyl imidazoles functioned as a tempered electrophile for assessing ligandability of select tyrosine and lysine sites in cell proteomes and when mated to a recognition element could produce targeted covalent inhibitors with reduced off-target activity. In summary, imidazole nucleofuges show balanced stability and tunability to produce sulfone-based electrophiles that bind functional tyrosine and lysine sites in the proteome.
Collapse
Affiliation(s)
- R Justin Grams
- Department of Chemistry, University of Texas at Austin, 100 E 24th St, Texas, 78712, United States
| | - Kun Yuan
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, 22904, United States
| | - Michael W Founds
- Department of Chemistry, University of Texas at Austin, 100 E 24th St, Texas, 78712, United States
| | - Madeleine L Ware
- Department of Chemistry, University of Texas at Austin, 100 E 24th St, Texas, 78712, United States
| | - Michael G Pilar
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, 22904, United States
| | - Ku-Lung Hsu
- Department of Chemistry, University of Texas at Austin, 100 E 24th St, Texas, 78712, United States
| |
Collapse
|
21
|
Xega V, Liu JL. Beyond reproduction: unraveling the impact of sex hormones on cardiometabolic health. MEDICAL REVIEW (2021) 2024; 4:284-300. [PMID: 39135604 PMCID: PMC11317208 DOI: 10.1515/mr-2024-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 05/07/2024] [Indexed: 08/15/2024]
Abstract
This review thoroughly explores the multifaceted roles of sexual hormones, emphasizing their impact beyond reproductive functions and underscoring their significant influence on cardiometabolic regulation. It analyzes the broader physiological implications of estrogen, testosterone, and progesterone, highlighting their effects on metabolic syndrome, lipid metabolism, glucose homeostasis, and cardiovascular health. Drawing from diverse molecular, clinical, and therapeutic studies, the paper delves into the intricate interplay between these hormones and cardiometabolic processes. By presenting a comprehensive analysis that goes beyond traditional perspectives, and recognizing sexual hormones as more than reproductive agents, the review sheds light on their broader significance in health and disease management, advocating for holistic and personalized medical approaches.
Collapse
Affiliation(s)
- Viktoria Xega
- MeDiC Program, The Research Institute of McGill University Health Centre, Montreal, Canada
| | - Jun-Li Liu
- Division of Endocrinology and Metabolism, Department of Medicine, McGill University, Montreal, Canada
| |
Collapse
|
22
|
Xiao J, Cao S, Wang J, Li P, Cheng Q, Zhou X, Dong J, Li Y, Zhao X, Xu Z, Yang L. Leptin-mediated suppression of lipoprotein lipase cleavage enhances lipid uptake and facilitates lymph node metastasis in gastric cancer. Cancer Commun (Lond) 2024; 44:855-878. [PMID: 38958445 PMCID: PMC11337928 DOI: 10.1002/cac2.12583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Lymph node metastasis (LNM) is the primary mode of metastasis in gastric cancer (GC). However, the precise mechanisms underlying this process remain elusive. Tumor cells necessitate lipid metabolic reprogramming to facilitate metastasis, yet the role of lipoprotein lipase (LPL), a pivotal enzyme involved in exogenous lipid uptake, remains uncertain in tumor metastasis. Therefore, the aim of this study was to investigate the presence of lipid metabolic reprogramming during LNM of GC as well as the role of LPL in this process. METHODS Intracellular lipid levels were quantified using oil red O staining, BODIPY 493/503 staining, and flow cytometry. Lipidomics analysis was employed to identify alterations in intracellular lipid composition following LPL knockdown. Protein expression levels were assessed through immunohistochemistry, Western blotting, and enzyme-linked immunosorbent assays. The mouse popliteal LNM model was utilized to investigate differences in LNM. Immunoprecipitation and mass spectrometry were employed to examine protein associations. In vitro phosphorylation assays and Phos-tag sodium dodecyl-sulfate polyacrylamide gel electrophoresis assays were conducted to detect angiopoietin-like protein 4 (ANGPTL4) phosphorylation. RESULTS We identified that an elevated intracellular lipid level represents a crucial characteristic of node-positive (N+) GC and further demonstrated that a high-fat diet can expedite LNM. LPL was found to be significantly overexpressed in N+ GC tissues and shown to facilitate LNM by mediating dietary lipid uptake within GC cells. Leptin, an obesity-related hormone, intercepted the effect exerted by ANGPTL4/Furin on LPL cleavage. Circulating leptin binding to the leptin receptor could induce the activation of inositol-requiring enzyme-1 (IRE1) kinase, leading to the phosphorylation of ANGPTL4 at the serine 30 residue and subsequently reducing its binding affinity with LPL. Moreover, our research revealed that LPL disrupted lipid homeostasis by elevating intracellular levels of arachidonic acid, which then triggered the cyclooxygenase-2/prostaglandin E2 (PGE2) pathway, thereby promoting tumor lymphangiogenesis. CONCLUSIONS Leptin-induced phosphorylation of ANGPTL4 facilitates LPL-mediated lipid uptake and consequently stimulates the production of PGE2, ultimately facilitating LNM in GC.
Collapse
Affiliation(s)
- Jian Xiao
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
| | - Shuqing Cao
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
| | - Jiawei Wang
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
| | - Pengyu Li
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
| | - Quan Cheng
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
| | - Xinyi Zhou
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
| | - Jiacheng Dong
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
| | - Yuan Li
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
| | - Xinyu Zhao
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
| | - Zekuan Xu
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
| | - Li Yang
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
| |
Collapse
|
23
|
Shang R, Rodrigues B. Lipoprotein lipase as a target for obesity/diabetes related cardiovascular disease. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:13199. [PMID: 39081272 PMCID: PMC11286490 DOI: 10.3389/jpps.2024.13199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 07/02/2024] [Indexed: 08/02/2024]
Abstract
Worldwide, the prevalence of obesity and diabetes have increased, with heart disease being their leading cause of death. Traditionally, the management of obesity and diabetes has focused mainly on weight reduction and controlling high blood glucose. Unfortunately, despite these efforts, poor medication management predisposes these patients to heart failure. One instigator for the development of heart failure is how cardiac tissue utilizes different sources of fuel for energy. In this regard, the heart switches from using various substrates, to predominantly using fatty acids (FA). This transformation to using FA as an exclusive source of energy is helpful in the initial stages of the disease. However, over the progression of diabetes this has grave end results. This is because toxic by-products are produced by overuse of FA, which weaken heart function (heart disease). Lipoprotein lipase (LPL) is responsible for regulating FA delivery to the heart, and its function during diabetes has not been completely revealed. In this review, the mechanisms by which LPL regulates fuel utilization by the heart in control conditions and following diabetes will be discussed in an attempt to identify new targets for therapeutic intervention. Currently, as treatment options to directly target diabetic heart disease are scarce, research on LPL may assist in drug development that exclusively targets fuel utilization by the heart and lipid accumulation in macrophages to help delay, prevent, or treat cardiac failure, and provide long-term management of this condition during diabetes.
Collapse
Affiliation(s)
- Rui Shang
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Brian Rodrigues
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
24
|
Wang J, Gu Y, Sun Y, Qiao Q, Huang X, Yang K, Bai Y. Adipogenic differentiation effect of human periodontal ligament stem cell initial cell density on autologous cells and human bone marrow stromal cells. Cell Biochem Funct 2024; 42:e4069. [PMID: 38940455 DOI: 10.1002/cbf.4069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/29/2024]
Abstract
Stem cells demonstrate differentiation and regulatory functions. In this discussion, we will explore the impacts of cell culture density on stem cell proliferation, adipogenesis, and regulatory abilities. This study aimed to investigate the impact of the initial culture density of human periodontal ligament stem cells (hPDLSCs) on the adipogenic differentiation of autologous cells. Our findings indicate that the proliferation rate of hPDLSCs increased with increasing initial cell density (0.5-8 × 104 cells/cm2). After adipogenic differentiation induced by different initial cell densities of hPDLSC, we found that the mean adipose concentration and the expression levels of lipoprotein lipase (LPL), CCAAT/enhancer binding protein α (CEBPα), and peroxisome proliferator-activated receptor γ (PPAR-γ) genes all increased with increasing cell density. To investigate the regulatory role of hPDLSCs in the adipogenic differentiation of other cells, we used secreted exocrine vesicles derived from hPDLSCs cultivated at different initial cell densities of 50 μg/mL to induce the adipogenic differentiation of human bone marrow stromal cells. We also found that the mean adipose concentration and expression of LPL, CEBPα, and PPARγ genes increased with increasing cell density, with an optimal culture density of 8 × 104 cells/cm2. This study provides a foundation for the application of adipogenic differentiation in stem cells.
Collapse
Affiliation(s)
- Jing Wang
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Yingzhi Gu
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Yaxi Sun
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Qingchen Qiao
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Xiaofeng Huang
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Kai Yang
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Yuxing Bai
- Department of Orthodontics, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
25
|
Zhang R, Zhang K. A unified model for regulating lipoprotein lipase activity. Trends Endocrinol Metab 2024; 35:490-504. [PMID: 38521668 PMCID: PMC11663433 DOI: 10.1016/j.tem.2024.02.016] [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: 12/25/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 03/25/2024]
Abstract
The regulation of triglyceride (TG) tissue distribution, storage, and utilization, a fundamental process of energy homeostasis, critically depends on lipoprotein lipase (LPL). We review the intricate mechanisms by which LPL activity is regulated by angiopoietin-like proteins (ANGPTL3, 4, 8), apolipoproteins (APOA5, APOC3, APOC2), and the cAMP-responsive element-binding protein H (CREBH). ANGPTL8 functions as a molecular switch, through complex formation, activating ANGPTL3 while deactivating ANGPTL4 in their LPL inhibition. The ANGPTL3-4-8 model integrates the roles of the aforementioned proteins in TG partitioning between white adipose tissue (WAT) and oxidative tissues (heart and skeletal muscles) during the feed/fast cycle. This model offers a unified perspective on LPL regulation, providing insights into TG metabolism, metabolic diseases, and therapeutics.
Collapse
Affiliation(s)
- Ren Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
26
|
Mirsalami SM, Mirsalami M. Evaluation of mesoporous silica particles as a support for lipase immobilization in biodiesel production: Enhanced ethyl ester synthesis from algal oil. RESULTS IN ENGINEERING 2024; 22:102138. [DOI: 10.1016/j.rineng.2024.102138] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
|
27
|
Chan DC, Watts GF. ANGPTL3 and ApoC-III inhibitors for treating hypertriglyceridemia in context: horses for courses? Curr Opin Lipidol 2024; 35:101-109. [PMID: 38372218 DOI: 10.1097/mol.0000000000000920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
PURPOSE OF REVIEW Hypertriglyceridemia (HTG) is an independent and casual risk factor for atherosclerotic cardiovascular disease (ASCVD). There is an unmet need for more effective treatments for patients with HTG. Angiopoietin-like protein 3 (ANGPTL3) and apolipoprotein C-III (apoC-III) are key regulators of triglyceride-rich lipoprotein (TRL) metabolism. We review recent clinical trials targeting ANGPTL3 and apoC-III with monoclonal antibody and nucleic acid therapies, including antisense oligonucleotides and small interfering RNA. RECENT FINDINGS ANGPTL3 and apoC-III inhibitors are effective in lowering plasma triglycerides and TRLs, with possibly greater efficacy with the inhibition of apoC-III. By contrast to ANGPTL3 inhibition that has the advantage of greater lowering of plasma low-density lipoprotein (LDL)-cholesterol and apoB levels, apoC-III inhibition only has a modest or no effect in lowering plasma LDL-cholesterol and apoB concentrations. Therapeutic inhibition of ANGPTL3 and apoC-III can correct HTG possibly by reducing production and increasing catabolism of TRL particles, but this remains to be formally investigated in patients with HTG. SUMMARY Novel agents targeting ANGPTL3 and apoC-III can correct HTG and potentially lower risk of ASCVD in patients with HTG. The long-term safety and cost-effectiveness of these agents await confirmation in ongoing and future studies.
Collapse
Affiliation(s)
- Dick C Chan
- Medical School, University of Western Australia
| | - Gerald F Watts
- Medical School, University of Western Australia
- Lipid Disorders Clinic, Department of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, Western Australia, Australia
| |
Collapse
|
28
|
Chandrasekaran P, Weiskirchen R. The signaling pathways in obesity-related complications. J Cell Commun Signal 2024; 18:e12039. [PMID: 38946722 PMCID: PMC11208128 DOI: 10.1002/ccs3.12039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 07/02/2024] Open
Abstract
Obesity, a rapidly expanding epidemic worldwide, is known to exacerbate many medical conditions, making it a significant factor in multiple diseases and their associated complications. This threatening epidemic is linked to various harmful conditions such as type 2 diabetes mellitus, hypertension, metabolic dysfunction-associated steatotic liver disease, polycystic ovary syndrome, cardiovascular diseases (CVDs), dyslipidemia, and cancer. The rise in urbanization and sedentary lifestyles creates an environment that fosters obesity, leading to both psychosocial and medical complications. To identify individuals at risk and ensure timely treatment, it is crucial to have a better understanding of the pathophysiology of obesity and its comorbidities. This comprehensive review highlights the relationship between obesity and obesity-associated complications, including type 2 diabetes, hypertension, (CVDs), dyslipidemia, polycystic ovary syndrome, metabolic dysfunction-associated steatotic liver disease, gastrointestinal complications, and obstructive sleep apnea. It also explores the potential mechanisms underlying these associations. A thorough analysis of the interplay between obesity and its associated complications is vital in developing effective therapeutic strategies to combat the exponential increase in global obesity rates and mitigate the deadly consequences of this polygenic condition.
Collapse
Affiliation(s)
| | - Ralf Weiskirchen
- Institute of Molecular PathobiochemistryExperimental Gene Therapy and Clinical Chemistry (IFMPEGKC)RWTH University Hospital AachenAachenGermany
| |
Collapse
|
29
|
Okamura T, Tsukamoto K, Arai H, Fujioka Y, Ishigaki Y, Koba S, Ohmura H, Shoji T, Yokote K, Yoshida H, Yoshida M, Deguchi J, Dobashi K, Fujiyoshi A, Hamaguchi H, Hara M, Harada-Shiba M, Hirata T, Iida M, Ikeda Y, Ishibashi S, Kanda H, Kihara S, Kitagawa K, Kodama S, Koseki M, Maezawa Y, Masuda D, Miida T, Miyamoto Y, Nishimura R, Node K, Noguchi M, Ohishi M, Saito I, Sawada S, Sone H, Takemoto M, Wakatsuki A, Yanai H. Japan Atherosclerosis Society (JAS) Guidelines for Prevention of Atherosclerotic Cardiovascular Diseases 2022. J Atheroscler Thromb 2024; 31:641-853. [PMID: 38123343 DOI: 10.5551/jat.gl2022] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Affiliation(s)
- Tomonori Okamura
- Preventive Medicine and Public Health, Keio University School of Medicine
| | | | | | - Yoshio Fujioka
- Faculty of Nutrition, Division of Clinical Nutrition, Kobe Gakuin University
| | - Yasushi Ishigaki
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University
| | - Shinji Koba
- Division of Cardiology, Department of Medicine, Showa University School of Medicine
| | - Hirotoshi Ohmura
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine
| | - Tetsuo Shoji
- Department of Vascular Medicine, Osaka Metropolitan University Graduate school of Medicine
| | - Koutaro Yokote
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine
| | - Hiroshi Yoshida
- Department of Laboratory Medicine, The Jikei University Kashiwa Hospital
| | | | - Juno Deguchi
- Department of Vascular Surgery, Saitama Medical Center, Saitama Medical University
| | - Kazushige Dobashi
- Department of Pediatrics, School of Medicine, University of Yamanashi
| | | | | | - Masumi Hara
- Department of Internal Medicine, Mizonokuchi Hospital, Teikyo University School of Medicine
| | - Mariko Harada-Shiba
- Cardiovascular Center, Osaka Medical and Pharmaceutical University
- Department of Molecular Pathogenesis, National Cerebral and Cardiovascular Center Research Institute
| | - Takumi Hirata
- Institute for Clinical and Translational Science, Nara Medical University
| | - Mami Iida
- Department of Internal Medicine and Cardiology, Gifu Prefectural General Medical Center
| | - Yoshiyuki Ikeda
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Jichi Medical University, School of Medicine
- Current affiliation: Ishibashi Diabetes and Endocrine Clinic
| | - Hideyuki Kanda
- Department of Public Health, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University
| | - Shinji Kihara
- Medical Laboratory Science and Technology, Division of Health Sciences, Osaka University graduate School of medicine
| | - Kazuo Kitagawa
- Department of Neurology, Tokyo Women's Medical University Hospital
| | - Satoru Kodama
- Department of Prevention of Noncommunicable Diseases and Promotion of Health Checkup, Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine
| | - Masahiro Koseki
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| | - Yoshiro Maezawa
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine
| | - Daisaku Masuda
- Department of Cardiology, Center for Innovative Medicine and Therapeutics, Dementia Care Center, Doctor's Support Center, Health Care Center, Rinku General Medical Center
| | - Takashi Miida
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine
| | | | - Rimei Nishimura
- Department of Diabetes, Metabolism and Endocrinology, The Jikei University School of Medicine
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University
| | - Midori Noguchi
- Division of Public Health, Department of Social Medicine, Graduate School of Medicine, Osaka University
| | - Mitsuru Ohishi
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University
| | - Isao Saito
- Department of Public Health and Epidemiology, Faculty of Medicine, Oita University
| | - Shojiro Sawada
- Division of Metabolism and Diabetes, Faculty of Medicine, Tohoku Medical and Pharmaceutical University
| | - Hirohito Sone
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine
| | - Minoru Takemoto
- Department of Diabetes, Metabolism and Endocrinology, International University of Health and Welfare
| | | | - Hidekatsu Yanai
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital
| |
Collapse
|
30
|
Zhu T, Zhao H, Chao Y, Gao S, Dong X, Wang Z. Olanzapine-induced weight gain and lipid dysfunction in mice between different gender. Biomed Chromatogr 2024; 38:e5864. [PMID: 38551083 DOI: 10.1002/bmc.5864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 02/27/2024] [Accepted: 03/03/2024] [Indexed: 05/21/2024]
Abstract
As one of the most common antipsychotics, olanzapine may cause metabolic-related adverse effects, but it is still unknown how olanzapine alters lipid metabolism. In this study, we found that olanzapine-treated mice showed varying degrees of dyslipidemia, which was particularly pronounced in female mice. Based on ultra-performance liquid chromatography-quadrupole time-of-flight-MS (UPLC-Q-TOF-MS) technology and lipid metabolomics, we mapped the changes in lipid metabolism in olanzapine-treated mice and then compared the changes in lipid metabolism between male and female mice. There were 98 metabolic differentiators between the olanzapine-treated and control groups in females and 79 in males. These metabolites were glycerolipids, glycerophospholipids, fatty amides, and sphingolipids, which are involved in glycerolipid metabolism, glycerophospholipid metabolism, and fatty acid metabolism. These results suggest that olanzapine-induced changes in the levels of lipid metabolites are closely associated with disturbances in lipid metabolic pathways, which may underlie lipemia. This lipidome profiling study not only visualizes changes in lipid metabolism in liver tissue but also provides a foundation for understanding the regulatory pathways and mechanisms involved in olanzapine-induced lipid metabolism disorders. Furthermore, this study demonstrates differences in lipid metabolism between males and females, providing a reference for clinical treatment regimen selection.
Collapse
Affiliation(s)
- Tong Zhu
- School of Medicine, Shanghai University, Shanghai, China
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Hongxia Zhao
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Zhanjiang, China
| | - Yufan Chao
- School of Medicine, Shanghai University, Shanghai, China
| | - Songyan Gao
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Xin Dong
- School of Medicine, Shanghai University, Shanghai, China
| | - Zuowei Wang
- School of Medicine, Shanghai University, Shanghai, China
- Division of Mood Disorders, Shanghai Hongkou Mental Health Center, Shanghai, China
- Clinical Research Center for Mental Health, School of Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
31
|
Xu D, Xie L, Cheng C, Xue F, Sun C. Triglyceride-rich lipoproteins and cardiovascular diseases. Front Endocrinol (Lausanne) 2024; 15:1409653. [PMID: 38883601 PMCID: PMC11176465 DOI: 10.3389/fendo.2024.1409653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 05/14/2024] [Indexed: 06/18/2024] Open
Abstract
The global prevalence of cardiovascular diseases (CVD) continues to rise steadily, making it a leading cause of mortality worldwide. Atherosclerosis (AS) serves as a primary driver of these conditions, commencing silently at an early age and culminating in adverse cardiovascular events that severely impact patients' quality of life or lead to fatality. Dyslipidemia, particularly elevated levels of low-density lipoprotein cholesterol (LDL-C), plays a pivotal role in AS pathogenesis as an independent risk factor. Research indicates that abnormal LDL-C accumulation within arterial walls acts as a crucial trigger for atherosclerotic plaque formation. As the disease progresses, plaque accumulation may rupture or dislodge, resulting in thrombus formation and complete blood supply obstruction, ultimately causing myocardial infarction, cerebral infarction, and other common adverse cardiovascular events. Despite adequate pharmacologic therapy targeting LDL-C reduction, patients with cardiometabolic abnormalities remain at high risk for disease recurrence, highlighting the importance of addressing lipid risk factors beyond LDL-C. Recent attention has focused on the causal relationship between triglycerides, triglyceride-rich lipoproteins (TRLs), and their remnants in AS risk. Genetic, epidemiologic, and clinical studies suggest a causal relationship between TRLs and their remnants and the increased risk of AS, and this dyslipidemia may be an independent risk factor for adverse cardiovascular events. Particularly in patients with obesity, metabolic syndrome, diabetes, and chronic kidney disease, disordered TRLs and its remnants levels significantly increase the risk of atherosclerosis and cardiovascular disease development. Accumulation of over-synthesized TRLs in plasma, impaired function of enzymes involved in TRLs lipolysis, and impaired hepatic clearance of cholesterol-rich TRLs remnants can lead to arterial deposition of TRLs and its remnants, promoting foam cell formation and arterial wall inflammation. Therefore, understanding the pathogenesis of TRLs-induced AS and targeting it therapeutically could slow or impede AS progression, thereby reducing cardiovascular disease morbidity and mortality, particularly coronary atherosclerotic heart disease.
Collapse
Affiliation(s)
- Dandan Xu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Lin Xie
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Cheng Cheng
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Fei Xue
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Chaonan Sun
- Department of Radiation Oncology, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang, China
| |
Collapse
|
32
|
Zupcic A, Latic N, Oubounyt M, Ramesova A, Carmeliet G, Baumbach J, Elkjaer ML, Erben RG. Ablation of Vitamin D Signaling in Cardiomyocytes Leads to Functional Impairment and Stimulation of Pro-Inflammatory and Pro-Fibrotic Gene Regulatory Networks in a Left Ventricular Hypertrophy Model in Mice. Int J Mol Sci 2024; 25:5929. [PMID: 38892126 PMCID: PMC11172934 DOI: 10.3390/ijms25115929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
The association between vitamin D deficiency and cardiovascular disease remains a controversial issue. This study aimed to further elucidate the role of vitamin D signaling in the development of left ventricular (LV) hypertrophy and dysfunction. To ablate the vitamin D receptor (VDR) specifically in cardiomyocytes, VDRfl/fl mice were crossed with Mlcv2-Cre mice. To induce LV hypertrophy experimentally by increasing cardiac afterload, transverse aortic constriction (TAC) was employed. Sham or TAC surgery was performed in 4-month-old, male, wild-type, VDRfl/fl, Mlcv2-Cre, and cardiomyocyte-specific VDR knockout (VDRCM-KO) mice. As expected, TAC induced profound LV hypertrophy and dysfunction, evidenced by echocardiography, aortic and cardiac catheterization, cardiac histology, and LV expression profiling 4 weeks post-surgery. Sham-operated mice showed no differences between genotypes. However, TAC VDRCM-KO mice, while having comparable cardiomyocyte size and LV fibrosis to TAC VDRfl/fl controls, exhibited reduced fractional shortening and ejection fraction as measured by echocardiography. Spatial transcriptomics of heart cryosections revealed more pronounced pro-inflammatory and pro-fibrotic gene regulatory networks in the stressed cardiac tissue niches of TAC VDRCM-KO compared to VDRfl/fl mice. Hence, our study supports the notion that vitamin D signaling in cardiomyocytes plays a protective role in the stressed heart.
Collapse
MESH Headings
- Animals
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Mice
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/pathology
- Receptors, Calcitriol/metabolism
- Receptors, Calcitriol/genetics
- Vitamin D/metabolism
- Gene Regulatory Networks
- Fibrosis
- Signal Transduction
- Male
- Disease Models, Animal
- Mice, Knockout
- Inflammation/metabolism
- Inflammation/genetics
- Inflammation/pathology
Collapse
Affiliation(s)
- Ana Zupcic
- Department of Biomedical Sciences, University of Veterinary Medicine, 1210 Vienna, Austria; (A.Z.); (N.L.); (A.R.)
| | - Nejla Latic
- Department of Biomedical Sciences, University of Veterinary Medicine, 1210 Vienna, Austria; (A.Z.); (N.L.); (A.R.)
| | - Mhaned Oubounyt
- Institute for Computational Systems Biology, University of Hamburg, Albert-Einstein-Ring 8-10, 22761 Hamburg, Germany; (J.B.); (M.L.E.)
| | - Alice Ramesova
- Department of Biomedical Sciences, University of Veterinary Medicine, 1210 Vienna, Austria; (A.Z.); (N.L.); (A.R.)
| | - Geert Carmeliet
- Department of Chronic Diseases, Metabolism and Ageing, 3000 Leuven, Belgium;
| | - Jan Baumbach
- Institute for Computational Systems Biology, University of Hamburg, Albert-Einstein-Ring 8-10, 22761 Hamburg, Germany; (J.B.); (M.L.E.)
| | - Maria L. Elkjaer
- Institute for Computational Systems Biology, University of Hamburg, Albert-Einstein-Ring 8-10, 22761 Hamburg, Germany; (J.B.); (M.L.E.)
| | - Reinhold G. Erben
- Department of Biomedical Sciences, University of Veterinary Medicine, 1210 Vienna, Austria; (A.Z.); (N.L.); (A.R.)
- Ludwig Boltzmann Institute of Osteology, Heinrich-Collin-Strasse 30, 1140 Vienna, Austria
| |
Collapse
|
33
|
Wu M, Wang Q, Li H, Tao J, Wang Z, Zhang S, Chen L, Li P, Chen L, Qu L. PLA2G12A protects against diet-induced obesity and insulin resistance by enhancing energy expenditure and clearance of circulating triglycerides. FASEB J 2024; 38:e23643. [PMID: 38703030 DOI: 10.1096/fj.202302075r] [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: 10/12/2023] [Revised: 03/11/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024]
Abstract
Secreted phospholipase A2s are involved in the development of obesity, type 2 diabetes mellitus (T2DM) and cardiovascular disease, which have become serious and growing health concerns worldwide. Integration of genome-wide association study and gene co-expression networks analysis showed that the secreted phospholipase A2 group XIIA (PLA2G12A) may participate in hepatic lipids metabolism. Nevertheless, the role of PLA2G12A in lipid metabolism and its potential mechanism remain elusive. Here, we used AAV9 vector carrying human PLA2G12A gene to exogenously express hPLA2G12A in the liver of mice. We demonstrated that the overexpression of hPLA2G12A resulted in a significant decrease in serum lipid levels in wild-type mice fed with chow diet or high-fat diet (HFD). Moreover, hPLA2G12A treatment protected against diet-induced obesity and insulin resistance in mice fed a HFD. Notably, we found that hPLA2G12A treatment confers protection against obesity and hyperlipidemia independent of its enzymatic activity, but rather by increasing physical activity and energy expenditure. Furthermore, we demonstrated that hPLA2G12A treatment induced upregulation of ApoC2 and Cd36 and downregulation of Angptl8, which contributed to the increase in clearance of circulating triglycerides and hepatic uptake of fatty acids without affecting hepatic de novo lipogenesis, very low-density lipoprotein secretion, or intestinal lipid absorption. Our study highlights the potential of PLA2G12A gene therapy as a promising approach for treating obesity, insulin resistance and T2DM.
Collapse
Affiliation(s)
- Min Wu
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qian Wang
- Guangzhou Laboratory, Guangzhou, China
| | - Hengchun Li
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiawang Tao
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhi Wang
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Shengnan Zhang
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Longhui Chen
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Pingchao Li
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ling Chen
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangzhou Laboratory, Guangzhou, China
| | - Linbing Qu
- Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
34
|
Alahmadi H, Martinez S, Farrell R, Bikienga R, Arinzeh N, Potts C, Li Z, Warner GR. Mixtures of phthalates disrupt expression of genes related to lipid metabolism and peroxisome proliferator-activated receptor signaling in mouse granulosa cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.02.592217. [PMID: 38746167 PMCID: PMC11092572 DOI: 10.1101/2024.05.02.592217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Phthalates are a class of known endocrine disrupting chemicals that are found in common everyday products. Several studies associate phthalate exposure with detrimental effects on ovarian functions, including growth and development of the follicle and production of steroid hormones. We hypothesized that dysregulation of the ovary by phthalates may be mediated by phthalate toxicity towards granulosa cells, a major cell type in ovarian follicles responsible for key steps of hormone production and nourishing the developing oocyte. To test the hypothesis that phthalates target granulosa cells, we harvested granulosa cells from adult CD-1 mouse ovaries and cultured them for 96 hours in vehicle control, a phthalate mixture, or a phthalate metabolite mixture (0.1-100 μg/mL). After culture, we measured metabolism of the phthalate mixture into monoester metabolites by the granulosa cells, finding that granulosa cells do not significantly contribute to ovarian metabolism of phthalates. Immunohistochemistry of phthalate metabolizing enzymes in whole ovaries confirmed that these enzymes are not strongly expressed in granulosa cells of antral follicles and that ovarian metabolism of phthalates likely occurs primarily in the stroma. RNA sequencing of treated granulosa cells identified 407 differentially expressed genes, with overrepresentation of genes from lipid metabolic processes, cholesterol metabolism, and peroxisome proliferator-activated receptor (PPAR) signaling pathways. Expression of significantly differentially expressed genes related to these pathways were confirmed using qPCR. Our results agree with previous findings that phthalates and phthalate metabolites have different effects on the ovary and interfere with PPAR signaling in granulosa cells.
Collapse
|
35
|
Gao N, Liu Y, Liu G, Liu B, Cheng Y. Sanghuangporus vaninii extract ameliorates hyperlipidemia in rats by mechanisms identified with transcriptome analysis. Food Sci Nutr 2024; 12:3360-3376. [PMID: 38726415 PMCID: PMC11077191 DOI: 10.1002/fsn3.4002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 01/18/2024] [Accepted: 01/21/2024] [Indexed: 05/12/2024] Open
Abstract
The increasing incidence of hyperlipidemia is a serious threat to public health. The development of effective and safe lipid-lowering drugs with few side effects is necessary. The purpose of this study was to assess the lipid-lowering activity of Sanghuangporus vaninii extract (SVE) in rat experiments and reveal the molecular mechanism by transcriptome analysis. Hyperlipidemia was induced in the animals using a high-fat diet for 4 weeks. At the end of the 4th week, hyperlipidemic rats were assigned into two control groups (model and positive simvastatin control) and three treatment groups that received SVE at 200, 400, or 800 mg kg-1 day-1 for another 4 weeks. A last control group comprised normal chow-fed rats. At the end of the 8th week, rats were sacrificed and lipid serum levels, histopathology, and liver transcriptome profiles were determined. SVE was demonstrated to relieve the lipid disorder and improve histopathological liver changes in a dose-dependent manner. The transcriptomic analysis identified changes in hepatocyte gene activity for major pathways including steroid biosynthesis, bile secretion, cholesterol metabolism, AMPK signaling, thyroid hormone signaling, and glucagon signaling. The changed expression of crucial genes in the different groups was confirmed by qPCR. Collectively, this study revealed that SVE could relieve hyperlipidemia in rats, the molecular mechanism might be to promote the metabolism of lipids and the excretion of cholesterol, inhibit the biosynthesis of cholesterol by activating the AMPK signaling pathway, the thyroid hormone signaling pathway, and the glucagon signaling pathway.
Collapse
Affiliation(s)
- Ning Gao
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of EducationHarbinChina
- School of PharmacyHeilongjiang University of Chinese MedicineHarbinChina
| | - Yuanzhen Liu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of EducationHarbinChina
- School of PharmacyHeilongjiang University of Chinese MedicineHarbinChina
| | - Guangjie Liu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of EducationHarbinChina
- School of PharmacyHeilongjiang University of Chinese MedicineHarbinChina
| | - Bo Liu
- School of Pharmaceutical EngineeringHeilongjiang Agricultural Reclamation Vocational CollegeHarbinChina
| | - Yupeng Cheng
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of EducationHarbinChina
- School of PharmacyHeilongjiang University of Chinese MedicineHarbinChina
| |
Collapse
|
36
|
Wen Y, Chen YQ, Konrad RJ. Angiopoietin-like protein 8: a multifaceted protein instrumental in regulating triglyceride metabolism. Curr Opin Lipidol 2024; 35:58-65. [PMID: 37962908 DOI: 10.1097/mol.0000000000000910] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
PURPOSE OF REVIEW The angiopoietin-like (ANGPTL) proteins ANGPTL3 and ANGPTL4 are critical lipoprotein lipase (LPL) inhibitors. This review discusses the unique ability of the insulin-responsive protein ANGPTL8 to regulate triglyceride (TG) metabolism by forming ANGPTL3/8 and ANGPTL4/8 complexes that control tissue-specific LPL activities. RECENT FINDINGS After feeding, ANGPTL4/8 acts locally in adipose tissue, has decreased LPL-inhibitory activity compared to ANGPTL4, and binds tissue plasminogen activator (tPA) and plasminogen to generate plasmin, which cleaves ANGPTL4/8 and other LPL inhibitors. This enables LPL to be fully active postprandially to promote efficient fatty acid (FA) uptake and minimize ectopic fat deposition. In contrast, liver-derived ANGPTL3/8 acts in an endocrine manner, has markedly increased LPL-inhibitory activity compared to ANGPTL3, and potently inhibits LPL in oxidative tissues to direct TG toward adipose tissue for storage. Circulating ANGPTL3/8 levels are strongly correlated with serum TG, and the ANGPTL3/8 LPL-inhibitory epitope is blocked by the TG-lowering protein apolipoprotein A5 (ApoA5). SUMMARY ANGPTL8 plays a crucial role in TG metabolism by forming ANGPTL3/8 and ANGPTL4/8 complexes that differentially modulate LPL activities in oxidative and adipose tissues respectively. Selective ANGPTL8 inhibition in the context of the ANGPTL3/8 complex has the potential to be a promising strategy for treating dyslipidemia.
Collapse
Affiliation(s)
- Yi Wen
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | | |
Collapse
|
37
|
Khodakarami Fard Z, Shirazinejad A, Mohammadi M, Hashemi SMB. Molecular Cloning of the Extracellular Lipases of Bacillus Amyloliquefaciens Isolated from Agrifood Wastes. IRANIAN JOURNAL OF BIOTECHNOLOGY 2024; 22:e3797. [PMID: 39220339 PMCID: PMC11364930 DOI: 10.30498/ijb.2024.417315.3797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 03/09/2024] [Indexed: 09/04/2024]
Abstract
Background The lipase enzyme (EC: 3.1.1.3) is one of the most important catalysts in food, dairy, detergent, and textile industries. Objective This study was performed to identify, isolate and characterize of lipase producing bacterial strain from agrifood wastes and to identify and characterize of their lipase genes. Materials and Methods In the present study, two lipase-producing isolates were identified from the effluent of Golbahar meat products and Soveyda vegetable oil factories using in silico and in vitro approaches. Results The results of morphological, biochemical, and molecular characterizations showed that both lipase-producing isolates belong to the Bacillus amyloliquefaciens species. Phylogenetic analysis confirmed the results of phenotypic, biochemical, and molecular characterizations. The results showed differences between LipA and LipB in the Golbahar and Soveyda isolates. Three different amino acids (residues 14, 100, and 165) were observed in LipA and one different amino acid (residue 102) was detected in LipB extracellular lipases. The protein molecular weight of the two extracted lipases ranged from 20 to 25 kDa. The identified extracellular lipases also had different physicochemical features. The maximum lipase activity of the Golbahar and Soveyda isolates was observed at 45 °C and at the pH of 8, but the Golbahar isolates exhibited higher lipase activity compared to the Soveyda isolates. The Golbahar and Soveyda isolates exhibited different activities in the presence of some ions, inhibitors, denaturing agents, and organic solvents and the Golbahar isolates showed better lipase activity than the Soveyda isolates. Conclusions In this study, two extracellular lipase-producing isolates of B. amyloliquefaciens were identified from different food industries, and their characteristics were investigated. The results of various investigations showed that the lipases produced by the Golbahar isolate have better characteristics than the lipases of the Soveyda isolate. The Golbahar lipases have a suitable temperature and pH activity range and maintain their activity in the presence of some ions, inhibitors, denaturing agents, and organic solvents. After further investigation, the Golbahar isolate lipase can be used in various industries. In addition, this lipase can be used enzyme engineering processes and its activity can be arbitrarily changed by targeted mutations. The results of this study can increase our knowledge of extracellular lipases and may turn out to have industrial applications.
Collapse
Affiliation(s)
- Zahra Khodakarami Fard
- Department of Food Science and Technology, Sarvestan Branch, Islamic Azad University, Sarvestan, Iran
| | - Alireza Shirazinejad
- Department of Food Science and Technology, Sarvestan Branch, Islamic Azad University, Sarvestan, Iran
| | - Mohsen Mohammadi
- Department of Pharmacognosy and Pharmaceutical Biotechnology, Faculty of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | | |
Collapse
|
38
|
Deng M, Kersten S. Characterization of sexual dimorphism in ANGPTL4 levels and function. J Lipid Res 2024; 65:100526. [PMID: 38431115 PMCID: PMC10973588 DOI: 10.1016/j.jlr.2024.100526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/05/2024] Open
Abstract
ANGPTL4 is an attractive pharmacological target for lowering plasma triglycerides and cardiovascular risk. Since most preclinical studies on ANGPTL4 were performed in male mice, little is known about sexual dimorphism in ANGPTL4 regulation and function. Here, we aimed to study potential sexual dimorphism in ANGPTL4 mRNA and protein levels and ANGPTL4 function. Additionally, we performed exploratory studies on the function of ANGPTL4 in the liver during fasting using Angptl4-transgenic and Angptl4-/- mice. Compared to female mice, male mice showed higher hepatic and adipose ANGPTL4 mRNA and protein levels, as well as a more pronounced effect of genetic ANGPTL4 modulation on plasma lipids. By contrast, very limited sexual dimorphism in ANGPTL4 levels was observed in human liver and adipose tissue. In human and mouse adipose tissue, ANGPTL8 mRNA and/or protein levels were significantly higher in females than males. Adipose LPL protein levels were higher in female than male Angptl4-/- mice, which was abolished by ANGPTL4 (over) expression. At the human genetic level, the ANGPTL4 E40K loss-of-function variant was associated with similar plasma triglyceride reductions in women and men. Finally, ANGPTL4 ablation in fasted mice was associated with changes in hepatic gene expression consistent with PPARα activation. In conclusion, the levels of ANGPTL4 and the magnitude of the effect of ANGPTL4 on plasma lipids exhibit sexual dimorphism. Nonetheless, inactivation of ANGPTL4 should confer a similar metabolic benefit in women and men. Expression levels of ANGPTL8 in human and mouse adipose tissue are highly sexually dimorphic, showing higher levels in females than males.
Collapse
Affiliation(s)
- Mingjuan Deng
- Nutrition, Metabolism, and Genomics group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Sander Kersten
- Nutrition, Metabolism, and Genomics group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands; Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
39
|
Packard CJ, Pirillo A, Tsimikas S, Ference BA, Catapano AL. Exploring apolipoprotein C-III: pathophysiological and pharmacological relevance. Cardiovasc Res 2024; 119:2843-2857. [PMID: 38039351 PMCID: PMC11484501 DOI: 10.1093/cvr/cvad177] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/21/2022] [Accepted: 02/07/2023] [Indexed: 12/03/2023] Open
Abstract
The availability of pharmacological approaches able to effectively reduce circulating LDL cholesterol (LDL-C) has led to a substantial reduction in the risk of atherosclerosis-related cardiovascular disease (CVD). However, a residual cardiovascular (CV) risk persists in treated individuals with optimal levels of LDL-C. Additional risk factors beyond LDL-C are involved, and among these, elevated levels of triglycerides (TGs) and TG-rich lipoproteins are causally associated with an increased CV risk. Apolipoprotein C-III (apoC-III) is a key regulator of TG metabolism and hence circulating levels through several mechanisms including the inhibition of lipoprotein lipase activity and alterations in the affinity of apoC-III-containing lipoproteins for both the hepatic receptors involved in their removal and extracellular matrix in the arterial wall. Genetic studies have clarified the role of apoC-III in humans, establishing a causal link with CVD and showing that loss-of-function mutations in the APOC3 gene are associated with reduced TG levels and reduced risk of coronary heart disease. Currently available hypolipidaemic drugs can reduce TG levels, although to a limited extent. Substantial reductions in TG levels can be obtained with new drugs that target specifically apoC-III; these include two antisense oligonucleotides, one small interfering RNA and an antibody.
Collapse
Affiliation(s)
- Chris J Packard
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Angela Pirillo
- Center for the Study of Atherosclerosis, E. Bassini Hospital, Milan, Italy
- Center for the Study of Dyslipidaemias, IRCCS MultiMedica, Sesto S. Giovanni, 20099 Milan, Italy
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, CA, USA
| | - Brian A Ference
- Centre for Naturally Randomized Trials, University of Cambridge, Cambridge, UK
| | - Alberico L Catapano
- Center for the Study of Dyslipidaemias, IRCCS MultiMedica, Sesto S. Giovanni, 20099 Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, via Balzaretti 9, 20133 Milan, Italy
| |
Collapse
|
40
|
Wu H, Xie J, Peng W, Ji F, Qian J, Shen Q, Hou G. Effects of guanidinoacetic acid supplementation on liver and breast muscle fat deposition, lipid levels, and lipid metabolism-related gene expression in ducks. Front Vet Sci 2024; 11:1364815. [PMID: 38435369 PMCID: PMC10904544 DOI: 10.3389/fvets.2024.1364815] [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: 01/03/2024] [Accepted: 02/02/2024] [Indexed: 03/05/2024] Open
Abstract
Exogenous supplementation of guanidinoacetic acid can mechanistically regulate the energy distribution in muscle cells. This study aimed to investigate the effects of guanidinoacetic acid supplementation on liver and breast muscle fat deposition, lipid levels, and lipid metabolism-related gene expression in ducks. We randomly divided 480 42 days-old female Jiaji ducks into four groups with six replicates and 20 ducks for each replicate. The control group was fed the basal diet, and the experimental groups were fed the basal diet with 400, 600, and 800 mg/kg (GA400, GA600, and GA800) guanidinoacetic acid, respectively. Compared with the control group, (1) the total cholesterol (p = 0.0262), triglycerides (p = 0.0357), malondialdehyde (p = 0.0452) contents were lower in GA400, GA600 and GA800 in the liver; (2) the total cholesterol (p = 0.0365), triglycerides (p = 0.0459), and malondialdehyde (p = 0.0326) contents in breast muscle were decreased in GA400, GA600 and GA800; (3) the high density lipoprotein (p = 0.0356) and apolipoprotein-A1 (p = 0.0125) contents were increased in GA600 in the liver; (4) the apolipoprotein-A1 contents (p = 0.0489) in breast muscle were higher in GA600 and GA800; (5) the lipoprotein lipase contents (p = 0.0325) in the liver were higher in GA600 and GA800; (6) the malate dehydrogenase contents (p = 0.0269) in breast muscle were lower in GA400, GA600, and GA800; (7) the insulin induced gene 1 (p = 0.0326), fatty acid transport protein 1 (p = 0.0412), and lipoprotein lipase (p = 0.0235) relative expression were higher in GA400, GA600, and GA800 in the liver; (8) the insulin induced gene 1 (p = 0.0269), fatty acid transport protein 1 (p = 0.0234), and lipoprotein lipase (p = 0.0425) relative expression were increased in GA400, GA600, and GA800 in breast muscle. In this study, the optimum dosage of 600 mg/kg guanidinoacetic acid improved the liver and breast muscle fat deposition, lipid levels, and lipid metabolism-related gene expression in ducks.
Collapse
Affiliation(s)
- Hongzhi Wu
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Jiajun Xie
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Weiqi Peng
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Fengjie Ji
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Jinyu Qian
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Qian Shen
- Hainan Xuhuai Technology Co., Ltd., Haikou, China
| | - Guanyu Hou
- Tropical Crop Genetic Resource Research Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| |
Collapse
|
41
|
Malinowski D, Safranow K, Pawlik A. LPL rs264, PROCR rs867186 and PDGF rs974819 Gene Polymorphisms in Patients with Unstable Angina. J Pers Med 2024; 14:213. [PMID: 38392646 PMCID: PMC10890678 DOI: 10.3390/jpm14020213] [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: 01/23/2024] [Revised: 02/11/2024] [Accepted: 02/14/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND Coronary artery disease is caused by changes in the coronary arteries due to the atherosclerotic process and thrombotic changes. A very important role in the development of the atherosclerotic process in the coronary vessels is played by the inflammatory process and the immune response. Due to the important role of lipids and the coagulation process in the atherosclerotic process, research has also focused on genes affecting lipid metabolism and the coagulation system. Lipoprotein lipase (LPL) is an enzyme that metabolises lipids, hydrolysing triglycerides to produce free fatty acids and glycerol. Protein C (PC) is an essential component of coagulation and fibrinolysis. It is activated on the endothelial surface by the membrane-bound thrombin-thrombomodulin complex. Platelet-derived growth factor (PDGF) has a number of important functions in processes related to fibroblast and smooth muscle cell function. Due to their influence on lipid metabolism and coagulation processes, LPL, PROCR (endothelial cell protein C receptor) and PDGF may affect the atherosclerotic process and, thus, the risk of coronary heart disease. The aim of the study was to examine the associations between the LPL rs264, PROCR rs867186 and PDGF rs974819 gene polymorphisms and the risk of unstable angina and selected clinical parameters. METHODS The study included 232 patients with unstable angina and 144 healthy subjects as the control group. Genotyping was performed using real-time PCR. RESULTS There were no statistically significant differences in the distribution of the polymorphisms tested between the patients with unstable angina and the control subjects. The results showed associations between the PROCR rs867186 and PDGF rs974819 polymorphisms and some clinical parameters in patients with unstable angina. In patients with the PDGF rs974819 CC genotype, there were increased values for cholesterol and LDL serum levels in comparison with patients with the PDGF rs974819 CT and TT genotypes. In patients with the PROCR rs867186 AA genotype, HDL serum levels were lower than in patients with the GA genotype. CONCLUSIONS The results of our study did not show that the LPL rs264, PROCR rs867186 and PDGF rs974819 gene polymorphisms were significant risk factors for unstable angina in our population. The results of the study suggest that PDGF rs974819 and PROCR rs867186 may be associated with some parameters of lipid metabolism.
Collapse
Affiliation(s)
- Damian Malinowski
- Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| |
Collapse
|
42
|
Zhao H, Liu X, Amantai X, Bi J, Cao X, Yue X. Characterization and Comparison Analysis of Milk Fat Globule Membrane Proteins between Human and Porcine Milk. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:3210-3217. [PMID: 38291649 DOI: 10.1021/acs.jafc.3c06298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
This study aimed to explore the differences in milk fat globule membrane (MFGM) proteins between human milk (HM) and porcine milk (PM) using a label-free quantitative proteomic approach. A total of 3920 and 4001 MFGM proteins were identified between PM and HM, respectively. Among them, 3520 common MFGM proteins were detected, including 956 significant differentially expressed MFGM proteins (DEPs). Gene ontology (GO) enrichment analysis showed that the DEPs were highly enriched in the lipid metabolic process and intrinsic component of membrane. Kyoto Encyclopedia of Genes and Genomes pathways suggested that protein processing in the endoplasmic reticulum was the most highly enriched pathway, followed by peroxisome, complement, and coagulation cascades. This study reflects the difference in the composition of MFGM proteins between HM and PM and provides a scientific and systematic reference for the development of MFGM protein nutrition.
Collapse
Affiliation(s)
- Huiwen Zhao
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Xiaoyu Liu
- Department of Obstetrics and Gynaecology, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Xiakouna Amantai
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Jiayang Bi
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Xueyan Cao
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Xiqing Yue
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| |
Collapse
|
43
|
Fortunato IM, Pereira QC, Oliveira FDS, Alvarez MC, dos Santos TW, Ribeiro ML. Metabolic Insights into Caffeine's Anti-Adipogenic Effects: An Exploration through Intestinal Microbiota Modulation in Obesity. Int J Mol Sci 2024; 25:1803. [PMID: 38339081 PMCID: PMC10855966 DOI: 10.3390/ijms25031803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/22/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Obesity, a chronic condition marked by the excessive accumulation of adipose tissue, not only affects individual well-being but also significantly inflates healthcare costs. The physiological excess of fat manifests as triglyceride (TG) deposition within adipose tissue, with white adipose tissue (WAT) expansion via adipocyte hyperplasia being a key adipogenesis mechanism. As efforts intensify to address this global health crisis, understanding the complex interplay of contributing factors becomes critical for effective public health interventions and improved patient outcomes. In this context, gut microbiota-derived metabolites play an important role in orchestrating obesity modulation. Microbial lipopolysaccharides (LPS), secondary bile acids (BA), short-chain fatty acids (SCFAs), and trimethylamine (TMA) are the main intestinal metabolites in dyslipidemic states. Emerging evidence highlights the microbiota's substantial role in influencing host metabolism and subsequent health outcomes, presenting new avenues for therapeutic strategies, including polyphenol-based manipulations of these microbial populations. Among various agents, caffeine emerges as a potent modulator of metabolic pathways, exhibiting anti-inflammatory, antioxidant, and obesity-mitigating properties. Notably, caffeine's anti-adipogenic potential, attributed to the downregulation of key adipogenesis regulators, has been established. Recent findings further indicate that caffeine's influence on obesity may be mediated through alterations in the gut microbiota and its metabolic byproducts. Therefore, the present review summarizes the anti-adipogenic effect of caffeine in modulating obesity through the intestinal microbiota and its metabolites.
Collapse
Affiliation(s)
- Isabela Monique Fortunato
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (I.M.F.); (Q.C.P.); (F.d.S.O.); (M.C.A.); (T.W.d.S.)
| | - Quélita Cristina Pereira
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (I.M.F.); (Q.C.P.); (F.d.S.O.); (M.C.A.); (T.W.d.S.)
| | - Fabricio de Sousa Oliveira
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (I.M.F.); (Q.C.P.); (F.d.S.O.); (M.C.A.); (T.W.d.S.)
| | - Marisa Claudia Alvarez
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (I.M.F.); (Q.C.P.); (F.d.S.O.); (M.C.A.); (T.W.d.S.)
- Hematology and Transfusion Medicine Center, University of Campinas/Hemocentro, UNICAMP, Rua Carlos Chagas 480, Campinas 13083-878, SP, Brazil
| | - Tanila Wood dos Santos
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (I.M.F.); (Q.C.P.); (F.d.S.O.); (M.C.A.); (T.W.d.S.)
| | - Marcelo Lima Ribeiro
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (I.M.F.); (Q.C.P.); (F.d.S.O.); (M.C.A.); (T.W.d.S.)
| |
Collapse
|
44
|
Papadopoulou P, van der Pol R, van Hilten N, van Os WL, Pattipeiluhu R, Arias-Alpizar G, Knol RA, Noteborn W, Moradi MA, Ferraz MJ, Aerts JMFG, Sommerdijk N, Campbell F, Risselada HJ, Sevink GJA, Kros A. Phase-Separated Lipid-Based Nanoparticles: Selective Behavior at the Nano-Bio Interface. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310872. [PMID: 37988682 DOI: 10.1002/adma.202310872] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Indexed: 11/23/2023]
Abstract
The membrane-protein interface on lipid-based nanoparticles influences their in vivo behavior. Better understanding may evolve current drug delivery methods toward effective targeted nanomedicine. Previously, the cell-selective accumulation of a liposome formulation in vivo is demonstrated, through the recognition of lipid phase-separation by triglyceride lipases. This exemplified how liposome morphology and composition can determine nanoparticle-protein interactions. Here, the lipase-induced compositional and morphological changes of phase-separated liposomes-which bear a lipid droplet in their bilayer- are investigated, and the mechanism upon which lipases recognize and bind to the particles is unravelled. The selective lipolytic degradation of the phase-separated lipid droplet is observed, while nanoparticle integrity remains intact. Next, the Tryptophan-rich loop of the lipase is identified as the region with which the enzymes bind to the particles. This preferential binding is due to lipid packing defects induced on the liposome surface by phase separation. In parallel, the existing knowledge that phase separation leads to in vivo selectivity, is utilized to generate phase-separated mRNA-LNPs that target cell-subsets in zebrafish embryos, with subsequent mRNA delivery and protein expression. Together, these findings can expand the current knowledge on selective nanoparticle-protein communications and in vivo behavior, aspects that will assist to gain control of lipid-based nanoparticles.
Collapse
Affiliation(s)
- Panagiota Papadopoulou
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Rianne van der Pol
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Niek van Hilten
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Winant L van Os
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Roy Pattipeiluhu
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Gabriela Arias-Alpizar
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Renzo Aron Knol
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Willem Noteborn
- NeCEN, Leiden University, Einsteinweg 55, Leiden, 2333 AL, The Netherlands
| | - Mohammad-Amin Moradi
- Department of Chemical Engineering and Chemistry, Eindhoven University of Technology, P. O. Box 513, Eindhoven, 5600 MB, The Netherlands
| | - Maria Joao Ferraz
- Department of Medical Biochemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | | | - Nico Sommerdijk
- Department of Chemical Engineering and Chemistry, Eindhoven University of Technology, P. O. Box 513, Eindhoven, 5600 MB, The Netherlands
- Department of Medical BioSciences and Radboud Technology Center - Electron Microscopy, Radboud University Medical Center, Nijmegen, 6525 GA, The Netherlands
| | - Frederick Campbell
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Herre Jelger Risselada
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
- Department of Physics, Technical University Dortmund, 44221, Dortmund, Germany
| | - Geert Jan Agur Sevink
- Department of Biophysical Organic Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| | - Alexander Kros
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P. O. Box 9502, Leiden, 2300 RA, The Netherlands
| |
Collapse
|
45
|
Keshvari S, Masson JJR, Ferrari-Cestari M, Bodea LG, Nooru-Mohamed F, Tse BWC, Sokolowski KA, Batoon L, Patkar OL, Sullivan MA, Ebersbach H, Stutz C, Parton RG, Summers KM, Pettit AR, Hume DA, Irvine KM. Reversible expansion of tissue macrophages in response to macrophage colony-stimulating factor (CSF1) transforms systemic lipid and carbohydrate metabolism. Am J Physiol Endocrinol Metab 2024; 326:E149-E165. [PMID: 38117267 DOI: 10.1152/ajpendo.00347.2023] [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: 10/26/2023] [Revised: 12/01/2023] [Accepted: 12/17/2023] [Indexed: 12/21/2023]
Abstract
Macrophages regulate metabolic homeostasis in health and disease. Macrophage colony-stimulating factor (CSF1)-dependent macrophages contribute to homeostatic control of the size of the liver. This study aimed to determine the systemic metabolic consequences of elevating circulating CSF1. Acute administration of a CSF1-Fc fusion protein to mice led to monocytosis, increased resident tissue macrophages in the liver and all major organs, and liver growth. These effects were associated with increased hepatic glucose uptake and extensive mobilization of body fat. The impacts of CSF1 on macrophage abundance, liver size, and body composition were rapidly reversed to restore homeostasis. The effects of CSF1 on metabolism were independent of several known endocrine regulators and did not impact the physiological fasting response. Analysis using implantable telemetry in metabolic cages revealed progressively reduced body temperature and physical activity with no change in diurnal food intake. These results demonstrate the existence of a dynamic equilibrium between CSF1, the mononuclear phagocyte system, and control of liver-to-body weight ratio, which in turn controls systemic metabolic homeostasis. This novel macrophage regulatory axis has the potential to promote fat mobilization, without changes in appetence, which may have novel implications for managing metabolic syndrome.NEW & NOTEWORTHY CSF1 administration expands tissue macrophages, which transforms systemic metabolism. CSF1 drives fat mobilization and glucose uptake to support liver growth. The effects of CSF1 are independent of normal hormonal metabolic regulation. The effects of CSF1 are rapidly reversible, restoring homeostatic body composition. CSF1-dependent macrophages and liver size are coupled in a dynamic equilibrium.
Collapse
Affiliation(s)
- Sahar Keshvari
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Jesse J R Masson
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Michelle Ferrari-Cestari
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Liviu-Gabriel Bodea
- Clem Jones Centre for Ageing and Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Fathima Nooru-Mohamed
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Brian W C Tse
- Preclinical Imaging Facility, Translational Research Institute, Brisbane, Queensland, Australia
| | - Kamil A Sokolowski
- Preclinical Imaging Facility, Translational Research Institute, Brisbane, Queensland, Australia
| | - Lena Batoon
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Omkar L Patkar
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Mitchell A Sullivan
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Hilmar Ebersbach
- Novartis Institutes for Biomedical Research (NIBR), Basel, Switzerland
| | - Cian Stutz
- Novartis Institutes for Biomedical Research (NIBR), Basel, Switzerland
| | - Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Queensland, Australia
| | - Kim M Summers
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Allison R Pettit
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - David A Hume
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Katharine M Irvine
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
46
|
Tate NM, Underwood M, Thomas-Hollands A, Minor KM, Cullen JN, Friedenberg SG, Mickelson JR, Xenoulis PG, Steiner JM, Furrow E. Sequence Analysis of Six Candidate Genes in Miniature Schnauzers with Primary Hypertriglyceridemia. Genes (Basel) 2024; 15:193. [PMID: 38397183 PMCID: PMC10888295 DOI: 10.3390/genes15020193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Miniature Schnauzers are predisposed to primary hypertriglyceridemia (HTG). In this study, we performed whole genome sequencing (WGS) of eight Miniature Schnauzers with primary HTG and screened for risk variants in six HTG candidate genes: LPL, APOC2, APOA5, GPIHBP1, LMF1, and APOE. Variants were filtered to identify those present in ≥2 Miniature Schnauzers with primary HTG and uncommon (<10% allele frequency) in a WGS variant database including 613 dogs from 61 other breeds. Three variants passed filtering: an APOE TATA box deletion, an LMF1 intronic SNP, and a GPIHBP1 missense variant. The APOE and GPIHBP1 variants were genotyped in a cohort of 108 Miniature Schnauzers, including 68 with primary HTG and 40 controls. A multivariable regression model, including age and sex, did not identify an effect of APOE (estimate = 0.18, std. error = 0.14; p = 0.20) or GPIHBP1 genotypes (estimate = -0.26, std. error = 0.42; p = 0.54) on triglyceride concentration. In conclusion, we did not identify a monogenic cause for primary HTG in Miniature Schnauzers in the six genes evaluated. However, if HTG in Miniature Schnauzers is a complex disease resulting from the cumulative effects of multiple variants and environment, the identified variants cannot be ruled out as contributing factors.
Collapse
Affiliation(s)
- Nicole M. Tate
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (K.M.M.); (S.G.F.); (E.F.)
| | - Michaela Underwood
- VCA Veterinary Specialty & Emergency Center of Kalamazoo, Kalamazoo, MI 49001, USA;
| | | | - Katie M. Minor
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (K.M.M.); (S.G.F.); (E.F.)
| | - Jonah N. Cullen
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA;
| | - Steven G. Friedenberg
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (K.M.M.); (S.G.F.); (E.F.)
| | - James R. Mickelson
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA;
| | - Panagiotis G. Xenoulis
- Clinic of Medicine, Faculty of Veterinary Medicine, University of Thessaly, 43100 Karditsa, Greece;
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, School of Veterinary and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA;
| | - Joerg M. Steiner
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, School of Veterinary and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA;
| | - Eva Furrow
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA; (K.M.M.); (S.G.F.); (E.F.)
| |
Collapse
|
47
|
Alves M, Laranjeira F, Correia-da-Silva G. Understanding Hypertriglyceridemia: Integrating Genetic Insights. Genes (Basel) 2024; 15:190. [PMID: 38397180 PMCID: PMC10887881 DOI: 10.3390/genes15020190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Hypertriglyceridemia is an exceptionally complex metabolic disorder characterized by elevated plasma triglycerides associated with an increased risk of acute pancreatitis and cardiovascular diseases such as coronary artery disease. Its phenotype expression is widely heterogeneous and heavily influenced by conditions as obesity, alcohol consumption, or metabolic syndromes. Looking into the genetic underpinnings of hypertriglyceridemia, this review focuses on the genetic variants in LPL, APOA5, APOC2, GPIHBP1 and LMF1 triglyceride-regulating genes reportedly associated with abnormal genetic transcription and the translation of proteins participating in triglyceride-rich lipoprotein metabolism. Hypertriglyceridemia resulting from such genetic abnormalities can be categorized as monogenic or polygenic. Monogenic hypertriglyceridemia, also known as familial chylomicronemia syndrome, is caused by homozygous or compound heterozygous pathogenic variants in the five canonical genes. Polygenic hypertriglyceridemia, also known as multifactorial chylomicronemia syndrome in extreme cases of hypertriglyceridemia, is caused by heterozygous pathogenic genetic variants with variable penetrance affecting the canonical genes, and a set of common non-pathogenic genetic variants (polymorphisms, using the former nomenclature) with well-established association with elevated triglyceride levels. We further address recent progress in triglyceride-lowering treatments. Understanding the genetic basis of hypertriglyceridemia opens new translational opportunities in the scope of genetic screening and the development of novel therapies.
Collapse
Affiliation(s)
- Mara Alves
- Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
| | - Francisco Laranjeira
- CGM—Centro de Genética Médica Jacinto de Magalhães, Centro Hospitalar Universitário de Santo António (CHUdSA), 4099-028 Porto, Portugal;
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-346 Porto, Portugal
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-600 Porto, Portugal
| | - Georgina Correia-da-Silva
- UCIBIO Applied Molecular Biosciences Unit and Associate Laboratory i4HB—Institute for Health and Bioeconomy Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
48
|
Wei X, Lu Y, Lin LL, Zhang C, Chen X, Wang S, Wu SA, Li ZJ, Quan Y, Sun S, Qi L. Proteomic screens of SEL1L-HRD1 ER-associated degradation substrates reveal its role in glycosylphosphatidylinositol-anchored protein biogenesis. Nat Commun 2024; 15:659. [PMID: 38253565 PMCID: PMC10803770 DOI: 10.1038/s41467-024-44948-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Endoplasmic reticulum-associated degradation (ERAD) plays indispensable roles in many physiological processes; however, the nature of endogenous substrates remains largely elusive. Here we report a proteomics strategy based on the intrinsic property of the SEL1L-HRD1 ERAD complex to identify endogenous ERAD substrates both in vitro and in vivo. Following stringent filtering using a machine learning algorithm, over 100 high-confidence potential substrates are identified in human HEK293T and mouse brown adipose tissue, among which ~88% are cell type-specific. One of the top shared hits is the catalytic subunit of the glycosylphosphatidylinositol (GPI)-transamidase complex, PIGK. Indeed, SEL1L-HRD1 ERAD attenuates the biogenesis of GPI-anchored proteins by specifically targeting PIGK for proteasomal degradation. Lastly, several PIGK disease variants in inherited GPI deficiency disorders are also SEL1L-HRD1 ERAD substrates. This study provides a platform and resources for future effort to identify proteome-wide endogenous substrates in vivo, and implicates SEL1L-HRD1 ERAD in many cellular processes including the biogenesis of GPI-anchored proteins.
Collapse
Affiliation(s)
- Xiaoqiong Wei
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - You Lu
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
- Life Sciences Institute and Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Liangguang Leo Lin
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Chengxin Zhang
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Xinxin Chen
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Siwen Wang
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Shuangcheng Alivia Wu
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Zexin Jason Li
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Yujun Quan
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
| | - Shengyi Sun
- Department of Pharmacology, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA
| | - Ling Qi
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, 22903, USA.
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA.
| |
Collapse
|
49
|
Danielewski M, Rapak A, Kruszyńska A, Małodobra-Mazur M, Oleszkiewicz P, Dzimira S, Kucharska AZ, Słupski W, Matuszewska A, Nowak B, Szeląg A, Piórecki N, Zaleska-Dorobisz U, Sozański T. Cornelian Cherry ( Cornus mas L.) Fruit Extract Lowers SREBP-1c and C/EBPα in Liver and Alters Various PPAR-α, PPAR-γ, LXR-α Target Genes in Cholesterol-Rich Diet Rabbit Model. Int J Mol Sci 2024; 25:1199. [PMID: 38256272 PMCID: PMC10816641 DOI: 10.3390/ijms25021199] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/13/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
Cornelian cherry (Cornus mas L.) fruits, abundant in iridoids and anthocyanins, are natural products with proven beneficial impacts on the functions of the cardiovascular system and the liver. This study aims to assess and compare whether and to what extent two different doses of resin-purified cornelian cherry extract (10 mg/kg b.w. or 50 mg/kg b.w.) applied in a cholesterol-rich diet rabbit model affect the levels of sterol regulatory element-binding protein 1c (SREBP-1c) and CCAAT/enhancer binding protein α (C/EBPα), and various liver X receptor-α (LXR-α), peroxisome proliferator-activated receptor-α (PPAR-α), and peroxisome proliferator-activated receptor-γ (PPAR-γ) target genes. Moreover, the aim is to evaluate the resistive index (RI) of common carotid arteries (CCAs) and aortas, and histopathological changes in CCAs. For this purpose, the levels of SREBP-1c, C/EBPα, ATP-binding cassette transporter A1 (ABCA1), ATP-binding cassette transporter G1 (ABCG1), fatty acid synthase (FAS), endothelial lipase (LIPG), carnitine palmitoyltransferase 1A (CPT1A), and adiponectin receptor 2 (AdipoR2) in liver tissue were measured. Also, the levels of lipoprotein lipase (LPL), visceral adipose tissue-derived serine protease inhibitor (Vaspin), and retinol-binding protein 4 (RBP4) in visceral adipose tissue were measured. The RI of CCAs and aortas, and histopathological changes in CCAs, were indicated. The oral administration of the cornelian cherry extract decreased the SREBP-1c and C/EBPα in both doses. The dose of 10 mg/kg b.w. increased ABCA1 and decreased FAS, CPT1A, and RBP4, and the dose of 50 mg/kg b.w. enhanced ABCG1 and AdipoR2. Mitigations in atheromatous changes in rabbits' CCAs were also observed. The obtained outcomes were compared to the results of our previous works. The beneficial results confirm that cornelian cherry fruit extract may constitute a potentially effective product in the prevention and treatment of obesity-related disorders.
Collapse
Affiliation(s)
- Maciej Danielewski
- Department of Pharmacology, Wroclaw Medical University, J. Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (W.S.); (A.M.); (B.N.); (A.S.)
| | - Andrzej Rapak
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland; (A.R.); (A.K.)
| | - Angelika Kruszyńska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland; (A.R.); (A.K.)
| | - Małgorzata Małodobra-Mazur
- Department of Forensic Medicine, Division of Molecular Techniques, Wroclaw Medical University, M. Sklodowskiej-Curie 52, 50-369 Wroclaw, Poland;
| | - Paweł Oleszkiewicz
- Department of Radiology and Imaging Diagnostics II, Lower Silesian Center of Oncology, Pulmonology and Hematology, Grabiszynska 105, 53-439 Wroclaw, Poland;
| | - Stanisław Dzimira
- Department of Pathology, Wroclaw University of Environmental and Life Sciences, C. K. Norwida 31, 50-375 Wroclaw, Poland;
| | - Alicja Z. Kucharska
- Department of Fruit, Vegetable, and Plant Nutraceutical Technology, Wroclaw University of Environmental and Life Sciences, J. Chelmonskiego 37, 51-630 Wroclaw, Poland;
| | - Wojciech Słupski
- Department of Pharmacology, Wroclaw Medical University, J. Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (W.S.); (A.M.); (B.N.); (A.S.)
| | - Agnieszka Matuszewska
- Department of Pharmacology, Wroclaw Medical University, J. Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (W.S.); (A.M.); (B.N.); (A.S.)
| | - Beata Nowak
- Department of Pharmacology, Wroclaw Medical University, J. Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (W.S.); (A.M.); (B.N.); (A.S.)
| | - Adam Szeląg
- Department of Pharmacology, Wroclaw Medical University, J. Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland; (W.S.); (A.M.); (B.N.); (A.S.)
| | - Narcyz Piórecki
- Bolestraszyce Arboretum and Institute of Physiography, Bolestraszyce 130, 37-722 Wyszatyce, Poland;
- Institute of Physical Culture Sciences, Medical College, University of Rzeszow, Cicha 2A, 35-326 Rzeszow, Poland
| | - Urszula Zaleska-Dorobisz
- Department of General and Pediatric Radiology, Wroclaw Medical University, M. Sklodowskiej-Curie 50/52, 50-369 Wroclaw, Poland;
| | - Tomasz Sozański
- Department of Preclinical Sciences, Pharmacology and Medical Diagnostics, Faculty of Medicine, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland;
| |
Collapse
|
50
|
Gugliucci A. The chylomicron saga: time to focus on postprandial metabolism. Front Endocrinol (Lausanne) 2024; 14:1322869. [PMID: 38303975 PMCID: PMC10830840 DOI: 10.3389/fendo.2023.1322869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/28/2023] [Indexed: 02/03/2024] Open
Abstract
Since statins have had such tremendous therapeutic success over the last three decades, the field of atherosclerosis has become somewhat LDL-centric, dismissing the relevance of triglycerides (TG), particularly chylomicrons, in atherogenesis. Nonetheless, 50% of patients who take statins are at risk of developing atherosclerotic cardiovascular disease (ASCVD) and are unable to achieve their goal LDL-C levels. This residual risk is mediated, in part by triglyceride rich lipoproteins (TRL) and their remnants. Following his seminal investigation on the subject, Zilversmit proposed that atherosclerosis is a postprandial event in 1979 (1-4). In essence, the concept suggests that remnant cholesterol-rich chylomicron (CM) and very-low density lipoprotein (VLDL) particles play a role in atherogenesis. Given the foregoing, this narrative review addresses the most recent improvements in our understanding of postprandial dyslipidemia. The primary metabolic pathways of chylomicrons are discussed, emphasizing the critical physiological role of lipoprotein lipase and apoCIII, the importance of these particles' fluxes in the postprandial period, their catabolic rate, the complexities of testing postprandial metabolism, and the role of angiopoietin-like proteins in the partition of CM during the fed cycle. The narrative is rounded out by the dysregulation of postprandial lipid metabolism in insulin resistance states and consequent CVD risk, the clinical evaluation of postprandial dyslipidemia, current research limits, and potential future study directions.
Collapse
Affiliation(s)
- Alejandro Gugliucci
- Glycation, Oxidation and Disease Laboratory, Department of Research, Touro University California, Vallejo, CA, United States
| |
Collapse
|