1
|
Wu CYC, Zhang Y, Howard P, Huang F, Lee RHC. ACSL3 is a promising therapeutic target for alleviating anxiety and depression in Alzheimer's disease. GeroScience 2025; 47:2383-2397. [PMID: 39532829 PMCID: PMC11978576 DOI: 10.1007/s11357-024-01424-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD), the leading cause of dementia, affects over 55 million people worldwide and is often accompanied by depression and anxiety. Both significantly impact patients' quality of life and impose substantial societal and economic burdens on healthcare systems. Identifying the complex regulatory mechanisms that contribute to the psychological and emotional deficits in AD will provide promising therapeutic targets. Biosynthesis of omega-3 (ω3) and omega-6 fatty acids (ω6-FA) through long-chain acyl-CoA synthetases (ACSL) is crucial for cell function and survival. This is due to ω3/6-FA's imperative role in modulating the plasma membrane, energy production, and inflammation. While ACSL dysfunction is known to cause heart, liver, and kidney diseases, the role of ACSL in pathological conditions in the central nervous system (e.g., depression and anxiety) remains largely unexplored. The impact of ACSLs on AD-related depression and anxiety was investigated in a mouse model of Alzheimer's disease (3xTg-AD). ACSL3 levels were significantly reduced in the hippocampus of aged 3xTg-AD mice (via capillary-based immunoassay). This reduction in ACAL3 was closely associated with increased depression and anxiety-like behavior (via forced swim, tail suspension, elevated plus maze, and sucrose preference test). Upregulation of ACSL3 via adenovirus in aged 3xTg-AD mice led to increased protein levels of brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor C (VEGF-C) (via brain histology, capillary-based immunoassay), resulting in alleviation of depression and anxiety symptoms. The present study highlights a novel neuroprotective role of ACSL3 in the brain. Targeting ACSL3 will offer an innovative approach for treating AD-related depression and anxiety.
Collapse
Affiliation(s)
- Celeste Yin-Chieh Wu
- Department of Neurology, Louisiana State University Health, LSU Health Sciences Center Shreveport, 1501 Kings Hwy, Shreveport, LA, 71103-3932, USA.
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA.
| | - Yulan Zhang
- Department of Neurology, Louisiana State University Health, LSU Health Sciences Center Shreveport, 1501 Kings Hwy, Shreveport, LA, 71103-3932, USA
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA
| | - Peyton Howard
- Department of Neurology, Louisiana State University Health, LSU Health Sciences Center Shreveport, 1501 Kings Hwy, Shreveport, LA, 71103-3932, USA
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA
| | - Fang Huang
- Department of Neurology, Louisiana State University Health, LSU Health Sciences Center Shreveport, 1501 Kings Hwy, Shreveport, LA, 71103-3932, USA
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA
| | - Reggie Hui-Chao Lee
- Department of Neurology, Louisiana State University Health, LSU Health Sciences Center Shreveport, 1501 Kings Hwy, Shreveport, LA, 71103-3932, USA
- Institute for Cerebrovascular and Neuroregeneration Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Cellular Biology and Anatomy, Louisiana State University Health, Shreveport, LA, USA
| |
Collapse
|
2
|
Noble JT, Bimpeh K, Pisciotta MA, Reyes Ballista JM, Hines KM, Brindley MA. Chikungunya Replication and Infection Is Dependent upon and Alters Cellular Hexosylceramide Levels in Vero Cells. Viruses 2025; 17:509. [PMID: 40284952 PMCID: PMC12031450 DOI: 10.3390/v17040509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/15/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
Chikungunya virus (CHIKV), a mosquito-borne alphavirus, causes significant global morbidity, including fever, rash, and persistent arthralgia. Utilizing untargeted lipidomics, we investigated how CHIKV infection alters host cell lipid metabolism in Vero cells. CHIKV infection induced marked catabolism of hexosylceramides, reducing their levels while increasing ceramide byproducts. Functional studies revealed a reliance on fatty acid synthesis, β-oxidation, and glycosphingolipid biosynthesis. Notably, inhibition of uridine diphosphate glycosyltransferase 8 (UGT8), essential for galactosylceramide production, significantly impaired CHIKV replication and entry in Vero cells. Sensitivity of CHIKV to UGT8 inhibition was reproduced in a disease-relevant cell line, mouse hepatocytes (Hepa1-6). CHIKV was also sensitive to evacetrapib, a cholesterol ester transfer protein (CETP) inhibitor, though the mechanism of inhibition appeared independent of CETP itself, suggesting an off-target effect. These findings highlight specific lipid pathways, particularly glycosphingolipid metabolism, as critical for CHIKV replication and further refine our understanding of how CHIKV exploits host lipid networks. This study provides new insights into CHIKV biology and suggests that targeted investigation of host lipid pathways may inform future therapeutic strategies.
Collapse
Affiliation(s)
- Joseph Thomas Noble
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (J.T.N.); (M.A.P.); (J.M.R.B.)
| | - Kingsley Bimpeh
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA; (K.B.); (K.M.H.)
| | - Michael Anthony Pisciotta
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (J.T.N.); (M.A.P.); (J.M.R.B.)
| | - Judith Mary Reyes Ballista
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (J.T.N.); (M.A.P.); (J.M.R.B.)
| | - Kelly Marie Hines
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA; (K.B.); (K.M.H.)
| | - Melinda Ann Brindley
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; (J.T.N.); (M.A.P.); (J.M.R.B.)
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
3
|
Guo Y, Jin C, Wei C, Zhong K, Gao Y, Li P, Qu Z, Bao Z, Wang B, Hu J. The Effects of Acute Temperature Changes on Transcriptomic Responses in the Liver of Leopard Coral Groupers ( Plectropomus leopardus). Antioxidants (Basel) 2025; 14:223. [PMID: 40002409 PMCID: PMC11851849 DOI: 10.3390/antiox14020223] [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: 11/27/2024] [Revised: 02/01/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
The leopard coral grouper (Plectropomus leopardus) is a commercially significant tropical marine species. With the ongoing effects of global climate change, increasing attention has been focused on leopard coral grouper's susceptibility to extreme cold weather. This study investigates the effects of acute cold exposure and temperature recovery on the liver of P. leopardus. Histological observations and enzyme activity assays revealed that temperature fluctuations caused significant disruptions to normal liver physiology, including lipid accumulation and alterations in antioxidant levels. Transcriptomic analysis of liver tissue identified 2744 differentially expressed genes (DEGs) across three experimental groups: 25 °C (control), 13 °C (cold exposure), and rewarming at 25 °C (R-25 °C). Functional enrichment analysis revealed that these DEGs were significantly associated with biological processes such as lipid metabolism and antioxidant defense, as well as pathways related to metabolism, fatty acid biosynthesis, and ferroptosis. Furthermore, dynamic regulation of lipid metabolism, immune responses, and oxidative stress pathways was observed in response to both cold stress and rewarming. Notably, several redox-related DEGs were identified, and their interactions with lipid metabolism were further explored. Additionally, representative DEGs associated with antioxidants and lipid metabolism, such as got1, gpx1a, gpt, and g6pcla.2, were validated by qRT-PCR and fluorescence in situ hybridization (FISH). Taken together, this study provides a systematic analysis of the effects of acute cold exposure and temperature recovery stress on the liver of the leopard coral grouper, laying the groundwork for further research on the temperature stress responses in teleost species.
Collapse
Affiliation(s)
- Yilan Guo
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Y.G.)
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya 572025, China
| | - Chaofan Jin
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Y.G.)
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya 572025, China
| | - Cun Wei
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Y.G.)
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya 572025, China
| | - Kangning Zhong
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Y.G.)
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya 572025, China
| | - Yurui Gao
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Y.G.)
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya 572025, China
| | - Peiyu Li
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Y.G.)
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya 572025, China
| | - Zhe Qu
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Y.G.)
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya 572025, China
| | - Zhenmin Bao
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Y.G.)
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya 572025, China
- Southern Marine Science and Engineer Guangdong Laboratory, Guangzhou 511458, China
| | - Bo Wang
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Y.G.)
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya 572025, China
| | - Jingjie Hu
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; (Y.G.)
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya 572025, China
- Southern Marine Science and Engineer Guangdong Laboratory, Guangzhou 511458, China
| |
Collapse
|
4
|
Ghosh S, Ganguly A, Habib M, Shin BC, Thamotharan S, Andersson S, Devaskar SU. Hepatic and Pancreatic Cellular Response to Early Life Nutritional Mismatch. Endocrinology 2025; 166:bqaf007. [PMID: 39823439 PMCID: PMC11815087 DOI: 10.1210/endocr/bqaf007] [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: 08/27/2024] [Revised: 11/22/2024] [Accepted: 01/15/2025] [Indexed: 01/19/2025]
Abstract
To determine the basis for perinatal nutritional mismatch causing metabolic dysfunction-associated steatotic liver disease and diabetes mellitus, we examined adult phenotype, hepatic transcriptome, and pancreatic β-islet function. In prenatal caloric-restricted rats with intrauterine growth restriction (IUGR) and postnatal exposure to high fat with fructose (HFhf) or high carbohydrate, we investigated male and female IUGR-HFhf and IUGR-high carbohydrate, vs HFhf and control offspring. Males more than females displayed adiposity, glucose intolerance, insulin resistance, hyperlipidemia, and hepatomegaly with hepatic steatosis. Male hepatic triglyceride synthesis, de novo lipogenesis genes increased, while female lipolysis, β-oxidation, fatty acid efflux, and FGF21 genes increased. IUGR-HFhf males demonstrated reduced β-islet insulin and humanin, and type 1 diabetes mellitus human amniotic fluid increased humanin. Humanin suppression disabled glucose stimulated insulin, ATP production, with apoptotic diminished β-islet viability. Humanin and FGF21 may reverse perinatal nutritional mismatched phenotype by restoring functional β islets and preventing metabolic dysfunction-associated steatotic liver disease and diabetes mellitus.
Collapse
Affiliation(s)
- Shubhamoy Ghosh
- Division of Neonatology & Developmental Biology, Department of Pediatrics, UCLA Children’s Discovery & Innovation Institute at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| | - Amit Ganguly
- Division of Neonatology & Developmental Biology, Department of Pediatrics, UCLA Children’s Discovery & Innovation Institute at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| | - Manal Habib
- Division of Endocrinology, Department of Pediatrics, UCLA Children’s Discovery & Innovation Institute at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| | - Bo-Chul Shin
- Division of Neonatology & Developmental Biology, Department of Pediatrics, UCLA Children’s Discovery & Innovation Institute at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| | - Shanthie Thamotharan
- Division of Neonatology & Developmental Biology, Department of Pediatrics, UCLA Children’s Discovery & Innovation Institute at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| | - Sture Andersson
- Department of Pediatrics, Helsinki University Central Hospital, 00290 Helsinki, Finland
| | - Sherin U Devaskar
- Division of Neonatology & Developmental Biology, Department of Pediatrics, UCLA Children’s Discovery & Innovation Institute at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1752, USA
| |
Collapse
|
5
|
Szymczak-Pajor I, Drzewoski J, Kozłowska M, Krekora J, Śliwińska A. The Gut Microbiota-Related Antihyperglycemic Effect of Metformin. Pharmaceuticals (Basel) 2025; 18:55. [PMID: 39861118 PMCID: PMC11768994 DOI: 10.3390/ph18010055] [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: 12/01/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
It is critical to sustain the diversity of the microbiota to maintain host homeostasis and health. Growing evidence indicates that changes in gut microbial biodiversity may be associated with the development of several pathologies, including type 2 diabetes mellitus (T2DM). Metformin is still the first-line drug for treatment of T2DM unless there are contra-indications. The drug primarily inhibits hepatic gluconeogenesis and increases the sensitivity of target cells (hepatocytes, adipocytes and myocytes) to insulin; however, increasing evidence suggests that it may also influence the gut. As T2DM patients exhibit gut dysbiosis, the intestinal microbiome has gained interest as a key target for metabolic diseases. Interestingly, changes in the gut microbiome were also observed in T2DM patients treated with metformin compared to those who were not. Therefore, the aim of this review is to present the current state of knowledge regarding the association of the gut microbiome with the antihyperglycemic effect of metformin. Numerous studies indicate that the reduction in glucose concentration observed in T2DM patients treated with metformin is due in part to changes in the biodiversity of the gut microbiota. These changes contribute to improved intestinal barrier integrity, increased production of short-chain fatty acids (SCFAs), regulation of bile acid metabolism, and enhanced glucose absorption. Therefore, in addition to the well-recognized reduction of gluconeogenesis, metformin also appears to exert its glucose-lowering effect by influencing gut microbiome biodiversity. However, we are only beginning to understand how metformin acts on specific microorganisms in the intestine, and further research is needed to understand its role in regulating glucose metabolism, including the impact of this remarkable drug on specific microorganisms in the gut.
Collapse
Affiliation(s)
- Izabela Szymczak-Pajor
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland;
| | - Józef Drzewoski
- Central Teaching Hospital of the Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland; (J.D.); (J.K.)
| | - Małgorzata Kozłowska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland;
| | - Jan Krekora
- Central Teaching Hospital of the Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland; (J.D.); (J.K.)
| | - Agnieszka Śliwińska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland;
| |
Collapse
|
6
|
Zhang W, Jiang Y, Ni Z, Zhou M, Liu L, Li X, Su S, Wang C. Identification of Copy Number Variations and Selection Signatures in Wannan Spotted Pigs by Whole Genome Sequencing Data: A Preliminary Study. Animals (Basel) 2024; 14:1419. [PMID: 38791637 PMCID: PMC11117326 DOI: 10.3390/ani14101419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Copy number variation (CNV) is an important structural variation used to elucidate complex economic traits. In this study, we sequenced 25 Wannan spotted pigs (WSPs) to detect their CNVs and identify their selection signatures compared with those of 10 Asian wild boars. A total of 14,161 CNVs were detected in the WSPs, accounting for 0.72% of the porcine genome. The fixation index (Fst) was used to identify the selection signatures, and 195 CNVs with the top 1% of the Fst value were selected. Eighty genes were identified in the selected CNV regions. Functional GO and KEGG analyses revealed that the genes within these selected CNVs are associated with key traits such as reproduction (GAL3ST1 and SETD2), fatty acid composition (PRKG1, ACACA, ACSL3, UGT8), immune system (LYZ), ear size (WIF1), and feed efficiency (VIPR2). The findings of this study contribute novel insights into the genetic CNVs underlying WSP characteristics and provide essential information for the protection and utilization of WSP populations.
Collapse
Affiliation(s)
- Wei Zhang
- Anhui Provincial Breeding Pig Genetic Evaluation Center, Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China; (W.Z.); (M.Z.); (L.L.); (X.L.)
| | - Yao Jiang
- National Animal Husbandry Service, Beijing 100125, China;
| | - Zelan Ni
- Anhui Provincial Livestock and Poultry Genetic Resources Conservation Center, Hefei 231283, China;
| | - Mei Zhou
- Anhui Provincial Breeding Pig Genetic Evaluation Center, Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China; (W.Z.); (M.Z.); (L.L.); (X.L.)
| | - Linqing Liu
- Anhui Provincial Breeding Pig Genetic Evaluation Center, Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China; (W.Z.); (M.Z.); (L.L.); (X.L.)
| | - Xiaoyu Li
- Anhui Provincial Breeding Pig Genetic Evaluation Center, Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China; (W.Z.); (M.Z.); (L.L.); (X.L.)
| | - Shiguang Su
- Anhui Provincial Breeding Pig Genetic Evaluation Center, Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China; (W.Z.); (M.Z.); (L.L.); (X.L.)
| | - Chonglong Wang
- Anhui Provincial Breeding Pig Genetic Evaluation Center, Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China; (W.Z.); (M.Z.); (L.L.); (X.L.)
| |
Collapse
|
7
|
Kobiela A, Hewelt-Belka W, Frąckowiak JE, Kordulewska N, Hovhannisyan L, Bogucka A, Etherington R, Piróg A, Dapic I, Gabrielsson S, Brown SJ, Ogg GS, Gutowska-Owsiak D. Keratinocyte-derived small extracellular vesicles supply antigens for CD1a-resticted T cells and promote their type 2 bias in the context of filaggrin insufficiency. Front Immunol 2024; 15:1369238. [PMID: 38585273 PMCID: PMC10995404 DOI: 10.3389/fimmu.2024.1369238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/07/2024] [Indexed: 04/09/2024] Open
Abstract
Introduction Exosome-enriched small extracellular vesicles (sEVs) are nanosized organelles known to participate in long distance communication between cells, including in the skin. Atopic dermatitis (AD) is a chronic inflammatory skin disease for which filaggrin (FLG) gene mutations are the strongest genetic risk factor. Filaggrin insufficiency affects multiple cellular function, but it is unclear if sEV-mediated cellular communication originating from the affected keratinocytes is also altered, and if this influences peptide and lipid antigen presentation to T cells in the skin. Methods Available mRNA and protein expression datasets from filaggrin-insufficient keratinocytes (shFLG), organotypic models and AD skin were used for gene ontology analysis with FunRich tool. sEVs secreted by shFLG and control shC cells were isolated from conditioned media by differential centrifugation. Mass spectrometry was carried out for lipidomic and proteomic profiling of the cells and sEVs. T cell responses to protein, peptide, CD1a lipid antigens, as well as phospholipase A2-digested or intact sEVs were measured by ELISpot and ELISA. Results Data analysis revealed extensive remodeling of the sEV compartment in filaggrin insufficient keratinocytes, 3D models and the AD skin. Lipidomic profiles of shFLGsEV showed a reduction in the long chain (LCFAs) and polyunsaturated fatty acids (PUFAs; permissive CD1a ligands) and increased content of the bulky headgroup sphingolipids (non-permissive ligands). This resulted in a reduction of CD1a-mediated interferon-γ T cell responses to the lipids liberated from shFLG-generated sEVs in comparison to those induced by sEVs from control cells, and an increase in interleukin 13 secretion. The altered sEV lipidome reflected a generalized alteration in the cellular lipidome in filaggrin-insufficient cells and the skin of AD patients, resulting from a downregulation of key enzymes implicated in fatty acid elongation and desaturation, i.e., enzymes of the ACSL, ELOVL and FADS family. Discussion We determined that sEVs constitute a source of antigens suitable for CD1a-mediated presentation to T cells. Lipids enclosed within the sEVs secreted on the background of filaggrin insufficiency contribute to allergic inflammation by reducing type 1 responses and inducing a type 2 bias from CD1a-restricted T cells, thus likely perpetuating allergic inflammation in the skin.
Collapse
Affiliation(s)
- Adrian Kobiela
- Laboratory of Experimental and Translational Immunology, Intercollegiate Faculty of Biotechnology of the University of Gdańsk and the Medical University of Gdańsk, Gdańsk, Poland
| | - Weronika Hewelt-Belka
- Department of Analytical Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Joanna E. Frąckowiak
- Laboratory of Experimental and Translational Immunology, Intercollegiate Faculty of Biotechnology of the University of Gdańsk and the Medical University of Gdańsk, Gdańsk, Poland
| | - Natalia Kordulewska
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury, Olsztyn, Poland
| | - Lilit Hovhannisyan
- Laboratory of Experimental and Translational Immunology, Intercollegiate Faculty of Biotechnology of the University of Gdańsk and the Medical University of Gdańsk, Gdańsk, Poland
| | - Aleksandra Bogucka
- The Mass Spectrometry Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Rachel Etherington
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Artur Piróg
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Irena Dapic
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Susanne Gabrielsson
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Sara J. Brown
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Graham S. Ogg
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Danuta Gutowska-Owsiak
- Laboratory of Experimental and Translational Immunology, Intercollegiate Faculty of Biotechnology of the University of Gdańsk and the Medical University of Gdańsk, Gdańsk, Poland
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
8
|
Wang W, Wang P, Zhu L, Liu B, Wei Q, Hou Y, Li X, Hu Y, Li W, Wang Y, Jiang C, Yang G, Wang J. An optimized fluorescent biosensor for monitoring long-chain fatty acyl-CoAs metabolism in vivo. Biosens Bioelectron 2024; 247:115935. [PMID: 38128319 DOI: 10.1016/j.bios.2023.115935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/07/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
Long-chain fatty acyl-CoAs (LCACoAs) are intermediates in lipid metabolism that exert a wide range of cellular functions. However, our knowledge about the subcellular distribution and regulatory impacts of LCACoAs is limited by a lack of methods for detecting LCACoAs in living cells and tissues. Here, we report our development of LACSerHR, a genetically encoded fluorescent biosensor that enables precise measurement of subtle fluctuations in the levels of endogenous LCACoAs in vivo. LACSerHR significantly improve the fluorescent brightness and analyte affinity, in vitro and in vivo testing showcased LACSerHR's large dynamic range. We demonstrate LACSerHR's capacity for real-time evaluation of LCACoA levels in specific subcellular compartments, for example in response to disruption of ACSL enzyme function in HEK293T cells. Moreover, we show the application of LACSerHR for sensitive measurement of elevated LCACoA levels in the livers of mouse models for two common metabolic diseases (NAFLD and type 2 diabetes). Thus, our LACSerHR sensor is a powerful, broadly applicable tool for studying LCACoAs metabolism and disease.
Collapse
Affiliation(s)
- Weibo Wang
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China; National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan, 430079, PR China
| | - Pengcheng Wang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, PR China
| | - Lixin Zhu
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Bingjie Liu
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Qingpeng Wei
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Yongkang Hou
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Xi Li
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Yufei Hu
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Wenzhe Li
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Yuan Wang
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, PR China
| | - Guangfu Yang
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan, 430079, PR China.
| | - Jing Wang
- State Key Laboratory of Natural and Biomimetic Drugs Department of Chemical Biology, School of Pharmaceutical Sciences Peking University, Beijing, 100191, PR China.
| |
Collapse
|
9
|
Wise JTF, Kondo K. Increased Lipogenesis Is Important for Hexavalent Chromium-Transformed Lung Cells and Xenograft Tumor Growth. Int J Mol Sci 2023; 24:17060. [PMID: 38069382 PMCID: PMC10707372 DOI: 10.3390/ijms242317060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
Hexavalent chromium, Cr(VI), is a known carcinogen and environmental health concern. It has been established that reactive oxygen species, genomic instability, and DNA damage repair deficiency are important contributors to the Cr(VI)-induced carcinogenesis mechanism. However, some hallmarks of cancer remain under-researched regarding the mechanism behind Cr(VI)-induced carcinogenesis. Increased lipogenesis is important to carcinogenesis and tumorigenesis in multiple types of cancers, yet the role increased lipogenesis has in Cr(VI) carcinogenesis is unclear. We report here that Cr(VI)-induced transformation of three human lung cell lines (BEAS-2B, BEP2D, and WTHBF-6) resulted in increased lipogenesis (palmitic acid levels), and Cr(VI)-transformed cells had an increased expression of key lipogenesis proteins (ATP citrate lyase [ACLY], acetyl-CoA carboxylase [ACC1], and fatty acid synthase [FASN]). We also determined that the Cr(VI)-transformed cells did not exhibit an increase in fatty acid oxidation or lipid droplets compared to their passage-matched control cells. Additionally, we observed increases in ACLY, ACC1, and FASN in lung tumor tissue compared with normal-adjacent lung tissue (in chromate workers that died of chromate-induced tumors). Next, using a known FASN inhibitor (C75), we treated Cr(VI)-transformed BEAS-2B with this inhibitor and measured cell growth, FASN protein expression, and growth in soft agar. We observed that FASN inhibition results in a decreased protein expression, decreased cell growth, and the inhibition of colony growth in soft agar. Next, using shRNA to knock down the FASN protein in Cr(VI)-transformed BEAS-2B cells, we saw a decrease in FASN protein expression and a loss of the xenograft tumor development of Cr(VI)-transformed BEAS-2B cells. These results demonstrate that FASN is important for Cr(VI)-transformed cell growth and cancer properties. In conclusion, these data show that Cr(VI)-transformation in vitro caused an increase in lipogenesis, and that this increase is vital for Cr(VI)-transformed cells.
Collapse
Affiliation(s)
- James T. F. Wise
- Wise Laboratory of Nutritional Toxicology and Metabolism, School of Nutrition and Food Sciences, College of Agriculture, Louisiana State University, 269 Knapp Hall, Baton Rouge, LA 70803, USA
- School of Nutrition and Food Sciences, College of Agriculture, Louisiana State University, Baton Rouge, LA 70803, USA
- School of Nutrition and Food Sciences, Louisiana State University Agriculture Center, Baton Rouge, LA 70803, USA
- Division of Nutritional Sciences, Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Kazuya Kondo
- Department of Oncological Medical Services, Graduate School of Biomedical Sciences, Tokushima University Graduate School, Tokushima City 770-8509, Japan
| |
Collapse
|
10
|
Ding N, Wang W, Teng J, Zeng Y, Zhang Q, Dong L, Tang H. miR-26a-5p Regulates Adipocyte Differentiation via Directly Targeting ACSL3 in Adipocytes. Adipocyte 2023; 12:1-10. [PMID: 36710425 PMCID: PMC9891161 DOI: 10.1080/21623945.2023.2166345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Preadipocytes become mature adipocytes after proliferation and differentiation, and although many genes and microRNAs have been identified in intramuscular fat, their physiological function and regulatory mechanisms remain largely unexplored. miR-26a-5p has been reported to be related to fat deposition, but its effect on porcine preadipocyte differentiation has not been explored. In this study, bioinformatics analysis and luciferase reporter assay identified that miR-26a-5p binds to the 3'UTR of Acyl-CoA synthetase long-chain family member 3 (ACSL3) mRNA. The model for porcine intramuscular preadipocyte differentiation was established to explore the function of miR-6a-5p-ACSL3 on adipocyte differentiation. ACSL3 knockdown markedly reduced the triglycerides (TG) content of cells, as well as the mRNA levels of adipogenic marker genes (PPAR-γ and SREBP-1c). The number of lipid droplets in cells transfected with a miR-26a-5p mimic is significantly reduced, consistent with ACSL3 knockdown results, while the miR-26a-5p inhibitor resulted in opposite results. Taken together, miR-26a-5p is a repressor of porcine preadipocyte differentiation and plays a vital role in ACSL3-mediated adipogenesis.
Collapse
Affiliation(s)
- Ning Ding
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science & Technology, Shandong Agricultural University, Taian, Shandong Province, China,Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian, China
| | - Wenwen Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science & Technology, Shandong Agricultural University, Taian, Shandong Province, China,Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian, China
| | - Jun Teng
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science & Technology, Shandong Agricultural University, Taian, Shandong Province, China,Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian, China
| | - Yongqing Zeng
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science & Technology, Shandong Agricultural University, Taian, Shandong Province, China,Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian, China
| | - Qin Zhang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science & Technology, Shandong Agricultural University, Taian, Shandong Province, China,Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian, China
| | - Licai Dong
- Shandong Futong Agriculture & Animal Husbandry Development Co. LTD, Linyi, China
| | - Hui Tang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science & Technology, Shandong Agricultural University, Taian, Shandong Province, China,Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian, China,CONTACT Hui Tang No. 61, Daizong Street, Tai’an City, Shandong Province, 271018, China
| |
Collapse
|
11
|
Zhao T, Zheng H, Xu JJ, Xu YC, Liu LL, Luo Z. MnO 2 nanoparticles and MnSO 4 differentially affected hepatic lipid metabolism through miR-92a/acsl3-dependent de novo lipogenesis in yellow catfish Pelteobagrusfulvidraco. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 336:122416. [PMID: 37598932 DOI: 10.1016/j.envpol.2023.122416] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 08/22/2023]
Abstract
With the increasing production and use of MnO2 NPs and MnSO4 in various fields, their discharge into the aquatic environment is inevitable, which poses potential threats to aquatic organisms and humans. However, to date, few studies have been conducted to investigate the potential mechanism of the toxicity of MnO2 NPs, and a comprehensive understanding of the differences between this mechanism and the toxicity mechanism of inorganic Mn (MnSO4) is still lacking. Since lipid metabolism-relevant parameters have been widely recognized as novel biomarkers for risk assessment of environmental contaminants, the present study investigated the differential mechanisms of how MnO2 NPs and MnSO4 affect hepatic lipid metabolism in a freshwater fish yellow catfish. Compared to MnSO4, dietary MnO2 NPs caused liver injury, increased hepatic lipid accumulation and induced lipotoxicity, and up-regulated mRNA expression of de novo lipogenic genes. Moreover, MnO2 NPs downregulated the expression of miR-92a and miR-92b-3p, microRNAs involved in regulation of lipid metabolism, in the liver. Mechanistically, we found that acls3, an acetyl-coenzyme A synthetase, is target gene of miR-92a, and miR-92a-acsl3-dependent de novo lipogenesis contributes to lipid accumulation and lipotoxicity induced by MnO2 NPs. Collectively, these findings provided novel insights into mechanism whereby miRNAs mediate nanoparticles- and inorganic Mn-induced hepatic lipotoxicity and changes of lipid metabolism in vertebrates. Our findings also shed new perspective for ecotoxicity and ecological risk of MnO2 NPs and MnSO4 in aquatic environment.
Collapse
Affiliation(s)
- Tao Zhao
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan, 430070, China
| | - Hua Zheng
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jie-Jie Xu
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yi-Chuang Xu
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan, 430070, China
| | - Lu-Lu Liu
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhi Luo
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agricultural University, Wuhan, 430070, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| |
Collapse
|
12
|
Uehara K, Santoleri D, Whitlock AEG, Titchenell PM. Insulin Regulation of Hepatic Lipid Homeostasis. Compr Physiol 2023; 13:4785-4809. [PMID: 37358513 PMCID: PMC10760932 DOI: 10.1002/cphy.c220015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
The incidence of obesity, insulin resistance, and type II diabetes (T2DM) continues to rise worldwide. The liver is a central insulin-responsive metabolic organ that governs whole-body metabolic homeostasis. Therefore, defining the mechanisms underlying insulin action in the liver is essential to our understanding of the pathogenesis of insulin resistance. During periods of fasting, the liver catabolizes fatty acids and stored glycogen to meet the metabolic demands of the body. In postprandial conditions, insulin signals to the liver to store excess nutrients into triglycerides, cholesterol, and glycogen. In insulin-resistant states, such as T2DM, hepatic insulin signaling continues to promote lipid synthesis but fails to suppress glucose production, leading to hypertriglyceridemia and hyperglycemia. Insulin resistance is associated with the development of metabolic disorders such as cardiovascular and kidney disease, atherosclerosis, stroke, and cancer. Of note, nonalcoholic fatty liver disease (NAFLD), a spectrum of diseases encompassing fatty liver, inflammation, fibrosis, and cirrhosis, is linked to abnormalities in insulin-mediated lipid metabolism. Therefore, understanding the role of insulin signaling under normal and pathologic states may provide insights into preventative and therapeutic opportunities for the treatment of metabolic diseases. Here, we provide a review of the field of hepatic insulin signaling and lipid regulation, including providing historical context, detailed molecular mechanisms, and address gaps in our understanding of hepatic lipid regulation and the derangements under insulin-resistant conditions. © 2023 American Physiological Society. Compr Physiol 13:4785-4809, 2023.
Collapse
Affiliation(s)
- Kahealani Uehara
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dominic Santoleri
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anna E. Garcia Whitlock
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paul M. Titchenell
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
13
|
Luo L, Liu Y, Nizigiyimana P, Ye M, Xiao Y, Guo Q, Su T, Luo X, Huang Y, Zhou H. DNA 6mA Demethylase ALKBH1 Orchestrates Fatty Acid Metabolism and Suppresses Diet-Induced Hepatic Steatosis. Cell Mol Gastroenterol Hepatol 2022; 14:1213-1233. [PMID: 36058506 PMCID: PMC9579408 DOI: 10.1016/j.jcmgh.2022.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/25/2022] [Accepted: 08/25/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND & AIMS Nonalcoholic fatty liver disease (NAFLD) is a major cause of liver-related morbidity and mortality whereas the pathogenic mechanism remains largely elusive. DNA N6-methyladenosine (6mA) modification is a recently identified epigenetic mark indicative of transcription in eukaryotic genomes. Here, we aimed to investigate the role and mechanism of DNA 6mA modification in NAFLD progression. METHODS Dot blot and immunohistochemistry were used to detect DNA 6mA levels. Liver-specific AlkB homolog 1 (ALKBH1)-knockout mice and mice with ALKBH1 overexpression in liver were subjected to a high-fat diet or methionine choline-deficient diet to evaluate the critical role of ALKBH1-demethylated DNA 6mA modification in the pathogenesis of hepatic steatosis during NAFLD. RNA sequencing and chromatin immunoprecipitation sequencing were performed to investigate molecular mechanisms underlying this process. RESULTS The DNA 6mA level was increased significantly with hepatic steatosis, while ALKBH1 expression was down-regulated markedly in both mouse and human fatty liver. Deletion of ALKBH1 in hepatocytes increased genomic 6mA levels and accelerated diet-induced hepatic steatosis and metabolic dysfunction. Comprehensive analyses of transcriptome and chromatin immunoprecipitation sequencing data indicated that ALKBH1 directly bound to and exclusively demethylated 6mA levels of genes involved in fatty acid uptake and lipogenesis, leading to reduced hepatic lipid accumulation. Importantly, ALKBH1 overexpression was sufficient to suppress lipid uptake and synthesis, and alleviated diet-induced hepatic steatosis and insulin resistance. CONCLUSIONS Our findings show an indispensable role of ALKBH1 as an epigenetic suppressor of DNA 6mA in hepatic fatty acid metabolism and offer a potential therapeutic target for NAFLD treatment.
Collapse
Affiliation(s)
- Liping Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ya Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Paul Nizigiyimana
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mingsheng Ye
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ye Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tian Su
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Haiyan Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China,Correspondence Address correspondence to: Haiyan Zhou, PhD, Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, No 87, Xiangya Road, Changsha, Hunan Province 410008, China.
| |
Collapse
|
14
|
Ma F, Zou Y, Ma L, Ma R, Chen X. Evolution, characterization, and immune response function of long-chain acyl-CoA synthetase genes in rainbow trout (Oncorhynchus mykiss) under hypoxic stress. Comp Biochem Physiol B Biochem Mol Biol 2022; 260:110737. [PMID: 35385771 DOI: 10.1016/j.cbpb.2022.110737] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/16/2022] [Accepted: 03/31/2022] [Indexed: 11/30/2022]
Abstract
Long-chain acyl-CoA synthetases (Acsls), members of the acyl-activating enzyme superfamily, haves been systematically characterized in mammals and certain fishes, but the research on their involvement in reproductive development and hypoxic stress response in rainbow trout remains limited. In this study, we investigated the acsl gene structure and physical and chemical characteristics and the evolutionary relationship among acsl genes using the NCBI/Ensembl database. Using hypoxia treatment experiment, acsl gene expression in various organs and its regulation were investigated. A total of 11 acsl genes were identified in rainbow trout. Phylogenetic analyses found that acsl genes in rainbow trout were clustered into two clades: acsl3/4 and acsl1/2/5/6, and the additional gene duplication observed resulted from the third round of genome duplication unique to teleosts. Multiple sequence alignment and conserved motif analyses showed that the sequence of acsl proteins was highly conserved. Real-time quantitative PCR (RT-qPCR) showed that the acsl genes were highly expressed in immune tissues (liver and head kidney). Under hypoxia, the expression of acsl genes was upregulated, suggesting that they enhance the tolerance to hypoxia and are involved in the immune response in rainbow trout. Our study provides valuable insights into teleost evolution and effects of hypoxia on biological immunity and form a basis for further research on the functional characteristics of acsl genes.
Collapse
Affiliation(s)
- Fang Ma
- Key Laboratory of Resource Utilization of Agricultural Solid Waste in Gansu Province, Tianshui Normal University, Tianshui, Gansu Province, PR China.
| | - Yali Zou
- Key Laboratory of Resource Utilization of Agricultural Solid Waste in Gansu Province, Tianshui Normal University, Tianshui, Gansu Province, PR China
| | - Langfang Ma
- Key Laboratory of Resource Utilization of Agricultural Solid Waste in Gansu Province, Tianshui Normal University, Tianshui, Gansu Province, PR China
| | - Ruilin Ma
- Key Laboratory of Resource Utilization of Agricultural Solid Waste in Gansu Province, Tianshui Normal University, Tianshui, Gansu Province, PR China
| | - Xin Chen
- Key Laboratory of Resource Utilization of Agricultural Solid Waste in Gansu Province, Tianshui Normal University, Tianshui, Gansu Province, PR China
| |
Collapse
|
15
|
Nguyen TH, Yousefi H, Okpechi SC, Lauterboeck L, Dong S, Yang Q, Alahari SK. Nischarin Deletion Reduces Oxidative Metabolism and Overall ATP: A Study Using a Novel NISCHΔ5-6 Knockout Mouse Model. Int J Mol Sci 2022; 23:ijms23031374. [PMID: 35163298 PMCID: PMC8835720 DOI: 10.3390/ijms23031374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 11/16/2022] Open
Abstract
Nischarin (Nisch) is a cytosolic scaffolding protein that harbors tumor-suppressor-like characteristics. Previous studies have shown that Nisch functions as a scaffolding protein and regulates multiple biological activities. In the current study, we prepared a complete Nisch knockout model, for the first time, by deletion of exons 5 and 6. This knockout model was confirmed by Qrt–PCR and Western blotting with products from mouse embryonic fibroblast (MEF) cells. Embryos and adult mice of knockouts are significantly smaller than their wild-type counterparts. Deletion of Nisch enhanced cell migration, as demonstrated by wound type and transwell migration assays. Since the animals were small in size, we investigated Nisch’s effect on metabolism by conducting several assays using the Seahorse analyzer system. These data indicate that Nisch null cells have lower oxygen consumption rates, lower ATP production, and lower levels of proton leak. We examined the expression of 15 genes involved in lipid and fat metabolism, as well as cell growth, and noted a significant increase in expression for many genes in Nischarin null animals. In summary, our results show that Nischarin plays an important physiological role in metabolic homeostasis.
Collapse
Affiliation(s)
- Tina H. Nguyen
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Science Center, New Orleans, LA 70112, USA; (T.H.N.); (H.Y.); (S.C.O.); (S.D.)
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Science Center, New Orleans, LA 70112, USA; (T.H.N.); (H.Y.); (S.C.O.); (S.D.)
| | - Samuel C. Okpechi
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Science Center, New Orleans, LA 70112, USA; (T.H.N.); (H.Y.); (S.C.O.); (S.D.)
| | - Lothar Lauterboeck
- Cardiovascular Center of Excellence, School of Medicine, Louisiana State University Health Science Center, New Orleans, LA 70112, USA; (L.L.); (Q.Y.)
- Department of Pharmacology, Louisiana State University Health Science Center, New Orleans, LA 70112, USA
| | - Shengli Dong
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Science Center, New Orleans, LA 70112, USA; (T.H.N.); (H.Y.); (S.C.O.); (S.D.)
| | - Qinglin Yang
- Cardiovascular Center of Excellence, School of Medicine, Louisiana State University Health Science Center, New Orleans, LA 70112, USA; (L.L.); (Q.Y.)
- Department of Pharmacology, Louisiana State University Health Science Center, New Orleans, LA 70112, USA
| | - Suresh K. Alahari
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Science Center, New Orleans, LA 70112, USA; (T.H.N.); (H.Y.); (S.C.O.); (S.D.)
- Correspondence: ; Tel.: +1-504-568-4734
| |
Collapse
|
16
|
Huang X, Gao Y, Cao H, Li J, Mo S, Li T, Wu J, Guo K, Wei R, Zhang K. Effects of scoparone on non-alcoholic fatty liver disease revealed by RNA sequencing. Front Endocrinol (Lausanne) 2022; 13:1004284. [PMID: 36157436 PMCID: PMC9500212 DOI: 10.3389/fendo.2022.1004284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 08/11/2022] [Indexed: 11/21/2022] Open
Abstract
Scoparone (SCO) is known to have curative effect of alleviating liver injury. The purpose of this study was to observe the therapeutic effect and possible mechanism of SCO against high-fat diet (HFD) induced non-alcoholic liver disease (NAFLD) through in vivo experiments and RNA sequencing. Male Kunming mice were fed with HFD for 8 weeks to establish a mouse model of NAFLD, and SCO was used to treat NAFLD. Histopathology and biochemical indicators were used to evaluate the liver injury and the efficacy of SCO. RNA sequencing analysis was performed to elucidate the hepatoprotective mechanism of SCO. Finally, the differentially expressed genes of cholesterol synthesis and fatty acid (triglyceride) synthesis pathways were verified by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. The histopathological results showed that HFD could lead to significant steatosis in mice, while SCO could alleviate liver steatosis remarkably in NAFLD mice. The determination of biochemical indicators showed that SCO could inhibit the increased serum transaminase activity and liver lipid level induced by HFD. RNA sequencing analysis of liver tissues found that 2742 and 3663 genes were significantly changed by HFD and SCO, respectively. SCO reversed the most of genes involved in cholesterol synthesis and fatty acid (triglyceride) metabolism induced by HFD. the results of the validation experiment were mostly consistent with the RNA sequencing. SCO alleviated liver injury and steatosis in NAFLD mice, which may be closely related to the regulation of cholesterol and fatty acid (triglyceride) metabolism.
Collapse
Affiliation(s)
- Xiaoyan Huang
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin, China
- Guangxi Key Laboratory of Diabetic Systems Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Ya Gao
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin, China
- Guangxi Key Laboratory of Diabetic Systems Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Houkang Cao
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin, China
| | - Jun Li
- Department of Obstetrics & Gynaecology, National University of Singapore, Singapore, Singapore
| | - Siyi Mo
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin, China
| | - Ting Li
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin, China
| | - Jianzhao Wu
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin, China
| | - Kai Guo
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin, China
| | - Riming Wei
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin, China
- Guangxi Key Laboratory of Diabetic Systems Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
- *Correspondence: Riming Wei, ; Kefeng Zhang,
| | - Kefeng Zhang
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin, China
- Guangxi Key Laboratory of Diabetic Systems Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
- *Correspondence: Riming Wei, ; Kefeng Zhang,
| |
Collapse
|
17
|
Ma Y, Nenkov M, Chen Y, Press AT, Kaemmerer E, Gassler N. Fatty acid metabolism and acyl-CoA synthetases in the liver-gut axis. World J Hepatol 2021; 13:1512-1533. [PMID: 34904027 PMCID: PMC8637682 DOI: 10.4254/wjh.v13.i11.1512] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/28/2021] [Accepted: 10/11/2021] [Indexed: 02/06/2023] Open
Abstract
Fatty acids are energy substrates and cell components which participate in regulating signal transduction, transcription factor activity and secretion of bioactive lipid mediators. The acyl-CoA synthetases (ACSs) family containing 26 family members exhibits tissue-specific distribution, distinct fatty acid substrate preferences and diverse biological functions. Increasing evidence indicates that dysregulation of fatty acid metabolism in the liver-gut axis, designated as the bidirectional relationship between the gut, microbiome and liver, is closely associated with a range of human diseases including metabolic disorders, inflammatory disease and carcinoma in the gastrointestinal tract and liver. In this review, we depict the role of ACSs in fatty acid metabolism, possible molecular mechanisms through which they exert functions, and their involvement in hepatocellular and colorectal carcinoma, with particular attention paid to long-chain fatty acids and small-chain fatty acids. Additionally, the liver-gut communication and the liver and gut intersection with the microbiome as well as diseases related to microbiota imbalance in the liver-gut axis are addressed. Moreover, the development of potentially therapeutic small molecules, proteins and compounds targeting ACSs in cancer treatment is summarized.
Collapse
Affiliation(s)
- Yunxia Ma
- Section Pathology, Institute of Forensic Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena 07747, Germany
| | - Miljana Nenkov
- Section Pathology, Institute of Forensic Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena 07747, Germany
| | - Yuan Chen
- Section Pathology, Institute of Forensic Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena 07747, Germany
| | - Adrian T Press
- Department of Anesthesiology and Intensive Care Medicine and Center for Sepsis Control and Care, Jena University Hospital, Friedrich Schiller University Jena, Jena 07747, Germany
| | - Elke Kaemmerer
- Department of Pediatrics, Jena University Hospital, Friedrich Schiller University Jena, Jena 07747, Germany
| | - Nikolaus Gassler
- Section Pathology, Institute of Forensic Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena 07747, Germany.
| |
Collapse
|
18
|
Suo L, Dai W, Chen X, Qin X, Li G, Song S, Zhang D, Zhang C. Proteomics analysis of N-methyl-d-aspartate-induced cell death in retinal and optic nerves. J Proteomics 2021; 252:104427. [PMID: 34781030 DOI: 10.1016/j.jprot.2021.104427] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 12/18/2022]
Abstract
This study aimed to comprehensively understand the proteomic characteristics and modulation of the neural microenvironment with N-methyl-d-aspartate (NMDA)-induced neuronal degeneration in the retina and optic nerve at 12 h after intravitreal injection of 40 nmol NMDA. Male Sprague-Dawley rats were sacrificed at 12 h after intravitreal injection of 40 nmol NMDA. PBS-injected eyes served as controls. The key cell death-linked proteins from the retina and optic nerve tissues were assessed by a mass spectrometry-based label-free approach. In proteomics analysis, we identified 3532 proteins in retinal tissues and 2593 proteins in optic nerve tissues. The ACSL3 (Q63151) and Prnp (P13852) proteins were upregulated in the NMDA-damaged retina and connected with ferroptosis. The Gabarapl2 (P60522) protein was upregulated in NMDA-damaged optic nerves and connected with autophagy. We performed parallel reaction monitoring (PRM) to validate the liquid chromatography-tandem mass spectrometry (LC-MS/MS) results. Data are available ProteomeXchange with identifiers PXD022466 (label-free quantification) and PXD022729 (PRM validation). SIGNIFICANCE: Excitotoxicity is one of the pathogeneses of various retinal disorders, including glaucoma, retinal ischemia-reperfusion and traumatic optic neuropathy. This study indicated that ferroptosis may be linked to pathological cell death in the retina with NMDA insult. Autophagy may be induced by NMDA overstimulation in both the optic nerve and retina. Regulating these types of death simultaneously may provide the maximum benefit for retinal disease therapy.
Collapse
Affiliation(s)
- Lingge Suo
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Wanwei Dai
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Xuhao Chen
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Xuejiao Qin
- Department of Ophthalmology, Shandong University Qilu Hospital, Shandong, China
| | - Guanlin Li
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Sijia Song
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Di Zhang
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Chun Zhang
- Department of Ophthalmology, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
19
|
Nan J, Lee JS, Lee SA, Lee DS, Park KS, Chung SS. An Essential Role of the N-Terminal Region of ACSL1 in Linking Free Fatty Acids to Mitochondrial β-Oxidation in C2C12 Myotubes. Mol Cells 2021; 44:637-646. [PMID: 34511469 PMCID: PMC8490201 DOI: 10.14348/molcells.2021.0077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/15/2021] [Accepted: 08/03/2021] [Indexed: 12/25/2022] Open
Abstract
Free fatty acids are converted to acyl-CoA by long-chain acyl-CoA synthetases (ACSLs) before entering into metabolic pathways for lipid biosynthesis or degradation. ACSL family members have highly conserved amino acid sequences except for their N-terminal regions. Several reports have shown that ACSL1, among the ACSLs, is located in mitochondria and mainly leads fatty acids to the β-oxidation pathway in various cell types. In this study, we investigated how ACSL1 was localized in mitochondria and whether ACSL1 overexpression affected fatty acid oxidation (FAO) rates in C2C12 myotubes. We generated an ACSL1 mutant in which the N-terminal 100 amino acids were deleted and compared its localization and function with those of the ACSL1 wild type. We found that ACSL1 adjoined the outer membrane of mitochondria through interaction of its N-terminal region with carnitine palmitoyltransferase-1b (CPT1b) in C2C12 myotubes. In addition, overexpressed ACSL1, but not the ACSL1 mutant, increased FAO, and ameliorated palmitate-induced insulin resistance in C2C12 myotubes. These results suggested that targeting of ACSL1 to mitochondria is essential in increasing FAO in myotubes, which can reduce insulin resistance in obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Jinyan Nan
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Ji Seon Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Seung-Ah Lee
- Genomic Medicine Institute, Seoul National University Medical Research Center, Seoul 03080, Korea
| | - Dong-Sup Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Kyong Soo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sung Soo Chung
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea
| |
Collapse
|
20
|
Identification of Long Non-Coding RNAs Involved in Porcine Fat Deposition Using Two High-Throughput Sequencing Methods. Genes (Basel) 2021; 12:genes12091374. [PMID: 34573356 PMCID: PMC8467702 DOI: 10.3390/genes12091374] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/30/2021] [Indexed: 12/13/2022] Open
Abstract
Adipose is an important body tissue in pigs, and fatty traits are critical in pig production. The function of long non-coding RNA (lncRNA) in fat deposition and metabolism has been found in previous studies. In this study, we collected the adipose tissue of six Landrace pigs with contrast backfat thickness (nhigh = 3, nlow = 3), after which we performed strand-specific RNA sequencing (RNA-seq) based on pooling and biological replicate methods. Biological replicate and pooling RNA-seq revealed 1870 and 1618 lncRNAs, respectively. Using edgeR, we determined that 1512 genes and 220 lncRNAs, 2240 genes and 127 lncRNAs were differentially expressed in biological replicate and pooling RNA-seq, respectively. After target gene prediction, we found that ACSL3 was cis-targeted by lncRNA TCONS-00052400 and could activate the conversion of long-chain fatty acids. In addition, lncRNA TCONS_00041740 cis-regulated gene ACACB regulated the rate-limiting enzyme in fatty acid oxidation. Since these genes have necessary functions in fat metabolism, the results imply that the lncRNAs detected in our study may affect backfat deposition in swine through regulation of their target genes. Our study explored the regulation of lncRNA and their target genes in porcine backfat deposition and provided new insights for further investigation of the biological functions of lncRNA.
Collapse
|
21
|
Chalil D, Aristizabal-Henao JJ, Chalil A, Stark KD. Evidence of multiple hepatic mechanisms to mobilize docosahexaenoic acid into dam plasma during pregnancy in chow-fed sprague dawley rats. Prostaglandins Leukot Essent Fatty Acids 2021; 171:102317. [PMID: 34245972 DOI: 10.1016/j.plefa.2021.102317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/17/2021] [Accepted: 06/17/2021] [Indexed: 11/16/2022]
Abstract
Fetal brain growth requires considerable amounts of docosahexaenoic acid (DHA) during late pregnancy that is associated with increased maternal/dam plasma levels of PC 16:0_22:6 (palmitoyl docosahexaenoyl phosphatidylcholine). While biosynthesis of DHA during pregnancy is upregulated, the mechanisms responsible for the incorporation of dam DHA into PC 16:0_22:6 are not understood. The present study used a discovery approach combining untargeted lipidomics of plasma and liver (n = 3/group) with semi-targeted qPCR of hepatic gene products (n = 6/group) to identify metabolic pathways related to DHA metabolism, with a hypothesis that an upregulated acyltransferase involved in PC remodeling would be identified. Sprague Dawley rats were fed a commercial rodent chow throughout the study and samples were collected before pregnancy (baseline), at 15 and 20 days of pregnancy, and 7 days postpartum. Plasma and hepatic PC 16:0_22:6 was significantly increased (by 79% and 194%, respectively) at day 20 of pregnancy. An increase in hepatic DG (diacylglycerol) 16:0_22:6 (by 243%) and significant decreases in Pla2G15 (0.4-fold) and Pla2G16 (0.6-fold) at day 20 of pregnancy, no changes in Lpcat1-4, and an abundant pool of hepatic pool PE (phosphatidylethanolamine) 16:0_22:6 suggest that plasma PC 16:0_22:6 is not being produced by fatty acyl remodeling during pregnancy. The increase in plasma PC 16:0_22:6 during pregnancy appears to be due to an increase in de novo synthesis of PC and both the CDP-choline and phosphatidylcholine methyltransferase pathways are implicated. There was also evidence suggesting channeling of DHA into PC and lipoprotein assembly may be occurring. Targeted research is necessary to confirm these findings, but the results of this study indicate metabolic adaptions to enable maternal/dam resiliency towards meeting the fetal/pup demand for DHA during pregnancy.
Collapse
Affiliation(s)
- Dan Chalil
- Department of Kinesiology, University of Waterloo, 200 University Avenue, Waterloo, ON, Canada, N2L 3G1
| | - Juan J Aristizabal-Henao
- Department of Kinesiology, University of Waterloo, 200 University Avenue, Waterloo, ON, Canada, N2L 3G1
| | - Alan Chalil
- Department of Kinesiology, University of Waterloo, 200 University Avenue, Waterloo, ON, Canada, N2L 3G1
| | - Ken D Stark
- Department of Kinesiology, University of Waterloo, 200 University Avenue, Waterloo, ON, Canada, N2L 3G1.
| |
Collapse
|
22
|
Immunohistochemical staining reveals differential expression of ACSL3 and ACSL4 in hepatocellular carcinoma and hepatic gastrointestinal metastases. Biosci Rep 2021; 40:222647. [PMID: 32286604 PMCID: PMC7198044 DOI: 10.1042/bsr20200219] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/24/2022] Open
Abstract
Long-chain fatty acyl CoA synthetases (ACSLs) activate fatty acids by CoA addition thus facilitating their intracellular metabolism. Dysregulated ACSL expression features in several cancers and can affect processes such as ferroptosis, fatty acid β-oxidation, prostaglandin biosynthesis, steroidogenesis and phospholipid acyl chain remodelling. Here we investigate long chain acyl-CoA synthetase 3 (ACSL3) and long chain acyl-CoA synthetase 4 (ACSL4) expression in liver malignancies. The expression and subcellular localisations of the ACSL3 and ACSL4 isoforms in hepatocellular carcinoma (HCC), cholangiocarcinoma (CCA) and hepatic metastases were assessed by immunohistochemical analyses of multiple tumour tissue arrays and by subcellular fractionation of cultured HepG2 cells. The expression of both enzymes was increased in HCC compared with normal liver. Expression of ACSL3 was similar in HCC and hepatic metastases but lower in healthy tissue. Increased ACSL3 expression distinguished HCC from CCA with a sensitivity of 87.2% and a specificity of 75%. ACSL4 expression was significantly greater in HCC than in all other tumours and distinguished HCC from normal liver tissue with a sensitivity of 93.8% and specificity of 93.6%. Combined ACSL3 and ACSL4 staining scores distinguished HCC from hepatic metastases with 80.1% sensitivity and 77.1% specificity. These enzymes had partially overlapping intracellular distributions, ACSL4 localised to the plasma membrane and both isoforms associated with lipid droplets and the endoplasmic reticulum (ER). In conclusion, analysis of ACSL3 and ACSL4 expression can distinguish different classes of hepatic tumours.
Collapse
|
23
|
Singh S, Nguyen HC, Ehsan M, Michels DCR, Singh P, Qadura M, Singh KK. Pravastatin-induced changes in expression of long non-coding and coding RNAs in endothelial cells. Physiol Rep 2021; 9:e14661. [PMID: 33369888 PMCID: PMC7769171 DOI: 10.14814/phy2.14661] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Atherosclerosis is the main cause of the cardiovascular disease (CVD). Elevated blood cholesterol and inflammation of the endothelium are two major mechanisms contributing to the establishment of atherosclerotic plaques. Statins, such as pravastatin, are blood-cholesterol lowering drugs commonly prescribed for patients with or at risk for CVDs. In addition to lowering blood cholesterols, statins have recently been shown to improve endothelial function in both hyper- and normocholesterolemic patients with atherosclerosis. To understand the molecular mechanisms underlying the endothelial function improvement by statins, we assessed the RNA profile of pravastatin-treated endothelial cells, particularly their mRNAs and long non-coding RNAs (lncRNAs). METHODS Human umbilical vein endothelial cells (HUVECs) treated with pravastatin (10 µM) for 24 hr were profiled for lncRNAs and mRNAs using the Arraystar Human lncRNA Expression Microarray V3.0. RESULTS Of the 30,584 different lncRNAs screened, 95 were significantly upregulated, while 86 were downregulated in HUVECs responding to pravastatin. LINC00281 and BC045663 were the most upregulated (~8-fold) and downregulated (~3.5-fold) lncRNAs, respectively. Of the 26,106 different mRNAs screened in the pravastatin-treated HUVEC samples, 190 were significantly upregulated, while 90 were downregulated. Assigning the differentially expressed genes by bioinformatics into functional groups revealed their molecular signaling involvement in the following physiological processes: osteoclast differentiation, Rap1 signaling pathway, hematopoiesis, immunity, and neurotrophin signaling pathway. CONCLUSIONS This is the first lncRNA and mRNA expression profiling of pravastatin-mediated changes in human endothelial cells. Our results reveal potential novel targets and mechanisms for pravastatin-mediated vascular protection in atherosclerosis.
Collapse
Affiliation(s)
- Shweta Singh
- Department of Chemical and Biochemical EngineeringSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Hien C. Nguyen
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
- Department of Anatomy and Cell BiologySchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Mehroz Ehsan
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
- Schulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - David C. R. Michels
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Priyanka Singh
- Schulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Mohammad Qadura
- Vascular SurgeryKeenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael’s HospitalTorontoONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
| | - Krishna K. Singh
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
- Department of Anatomy and Cell BiologySchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
- Pharmacology and ToxicologyUniversity of TorontoTorontoONCanada
| |
Collapse
|
24
|
Fernandez RF, Ellis JM. Acyl-CoA synthetases as regulators of brain phospholipid acyl-chain diversity. Prostaglandins Leukot Essent Fatty Acids 2020; 161:102175. [PMID: 33031993 PMCID: PMC8693597 DOI: 10.1016/j.plefa.2020.102175] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/22/2020] [Accepted: 09/09/2020] [Indexed: 12/20/2022]
Abstract
Each individual cell-type is defined by its distinct morphology, phenotype, molecular and lipidomic profile. The importance of maintaining cell-specific lipidomic profiles is exemplified by the numerous diseases, disorders, and dysfunctional outcomes that occur as a direct result of altered lipidome. Therefore, the mechanisms regulating cellular lipidome diversity play a role in maintaining essential biological functions. The brain is an organ particularly rich in phospholipids, the main constituents of cellular membranes. The phospholipid acyl-chain profile of membranes in the brain is rather diverse due in part to the high degree of cellular heterogeneity. These membranes and the acyl-chain composition of their phospholipids are highly regulated, but the mechanisms that confer this tight regulation are incompletely understood. A family of enzymes called acyl-CoA synthetases (ACSs) stands at a pinnacle step allowing influence over cellular acyl-chain selection and subsequent metabolic flux. ACSs perform the initial reaction for cellular fatty acid metabolism by ligating a Coenzyme A to a fatty acid which both traps a fatty acid within a cell and activates it for metabolism. The ACS family of enzymes is large and diverse consisting of 25-26 family members that are nonredundant, each with unique distribution across and within cell types, and differential fatty acid substrate preferences. Thus, ACSs confer a critical intracellular fatty acid selecting step in a cell-type dependent manner providing acyl-CoA moieties that serve as essential precursors for phospholipid synthesis and remodeling, and therefore serve as a key regulator of cellular membrane acyl-chain compositional diversity. Here we will discuss how the contribution of individual ACSs towards brain lipid metabolism has only just begun to be elucidated and discuss the possibilities for how ACSs may differentially regulate brain lipidomic diversity.
Collapse
Affiliation(s)
- Regina F Fernandez
- Department of Physiology and East Carolina Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, NC, United States
| | - Jessica M Ellis
- Department of Physiology and East Carolina Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, NC, United States.
| |
Collapse
|
25
|
Quercetin Improving Lipid Metabolism by Regulating Lipid Metabolism Pathway of Ileum Mucosa in Broilers. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8686248. [PMID: 33014279 PMCID: PMC7520004 DOI: 10.1155/2020/8686248] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/28/2020] [Accepted: 08/29/2020] [Indexed: 02/06/2023]
Abstract
This study is aimed at evaluating the regulatory mechanism of quercetin on lipid metabolism in the ileum of broilers to better understand these pathways decreasing abdominal fat. 480 chickens were randomly divided into 4 groups (control, 0.02% quercetin, 0.04% quercetin, and 0.06% quercetin). Breast muscle, thigh muscle, and abdominal fat pad were removed and weighed at 42 d of age. Serum was obtained by centrifuging blood samples from the jugular vein (10 ml) to determine high-density lipoprotein (HDL), total cholesterol (TC), low-density lipoprotein (LDL), triglyceride (TG), leptin, and adiponectin using ELISA. About 5 g of the ileum was harvested and immediately frozen in liquid nitrogen for RNA-seq. Then, the confirmation of RNA-seq results by the Real-Time Quantitative PCR (RT-qPCR) method was evaluated using Pearson's correlation. Compared with control, abdominal fat percentage was significantly decreased with increasing quercetin supplementation, and the best result was obtained at 0.06% dietary quercetin supplementation (P < 0.01). Breast muscle percentage was significantly decreased at 0.02% quercetin (P < 0.01), and thigh muscle percentage tended to increase (P = 0.078). Meanwhile, 0.04% and 0.06% quercetin significantly decreased TG (P < 0.01), TC (P < 0.01), and LDL content (P < 0.05) in serum. Serum leptin and adiponectin contents were significantly increased by 0.04% and 0.06% dietary quercetin supplementation, compared with the control (P < 0.01). Analyses of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) database were used to identify differently expressed genes and lipid metabolism pathways. Quercetin decreased abdominal fat percentage through regulating fat digestion and absorption, glycerophospholipid metabolism, AMPK signaling pathway, fatty acid degradation, and cholesterol metabolism.
Collapse
|
26
|
Guo J, Mo J, Zhao Q, Han Q, Kanerva M, Iwata H, Li Q. De novo transcriptomic analysis predicts the effects of phenolic compounds in Ba River on the liver of female sharpbelly (Hemiculter lucidus). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 264:114642. [PMID: 32408079 DOI: 10.1016/j.envpol.2020.114642] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/18/2020] [Accepted: 04/19/2020] [Indexed: 06/11/2023]
Abstract
This work aimed at predicting the toxic effects of phenolic compounds in Ba River on the health of female sharpbelly (Hemiculter lucidus) by the de novo transcriptomic analysis of the liver. Sharpbelly, a native fish living in freshwater ecosystem of East Asia, were sampled upstream, near, and downstream of a wastewater discharge to the Ba river. Based on the occurrence of bisphenol A (BPA), nonylphenol (NP), and 4-tert-octylphenol (4-t-OP) in the water and fish sampled from each site, up-, mid-, and down-stream were interpreted as control, high, and low treatment groups, respectively. In the mid-stream group the Fulton's condition factor (CF) and body weight were remarkably increased by approximate 20%; the gonado-somatic index (GSI) and hepatosomatic index (HSI) in mid-stream fish showed a similar increasing trend but lacking of statistical difference. Exposure to wastewater effluent caused 160 and 162 differentially expressed genes (DEGs) in up-mid and down-mid stream groups, respectively. Two sets of DEGs were primarily enriched in the signaling pathways of drug metabolism, endocrine system, cellular process, and lipid metabolism in the mid-stream sharpbelly, which may alter the fish behavior, disrupt the reproductive function, and lead to hypothyroidism, hepatic steatosis, etc. Taken together, our results linked the disrupted signaling pathways with activities of phenolic compounds to predict the potential effects of wastewater effluent on the health of wild fish.
Collapse
Affiliation(s)
- Jiahua Guo
- Shaanxi Key Laboratory of Earth Surface System and Environmental Carrying Capacity, College of Urban and Environmental Sciences, Northwest University, Xi'an, 710127, China
| | - Jiezhang Mo
- Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Qian Zhao
- Shaanxi Key Laboratory of Earth Surface System and Environmental Carrying Capacity, College of Urban and Environmental Sciences, Northwest University, Xi'an, 710127, China
| | - Qizhi Han
- Shaanxi Key Laboratory of Earth Surface System and Environmental Carrying Capacity, College of Urban and Environmental Sciences, Northwest University, Xi'an, 710127, China
| | - Mirella Kanerva
- Center for Marine Environmental Studies, Ehime University, Bunkyo-cho 2-5, Matsuyama, Ehime prefecture, 790-8577, Japan
| | - Hisato Iwata
- Center for Marine Environmental Studies, Ehime University, Bunkyo-cho 2-5, Matsuyama, Ehime prefecture, 790-8577, Japan
| | - Qi Li
- Shaanxi Key Laboratory of Earth Surface System and Environmental Carrying Capacity, College of Urban and Environmental Sciences, Northwest University, Xi'an, 710127, China.
| |
Collapse
|
27
|
Jung YH, Bu SY. Suppression of long chain acyl-CoA synthetase blocks intracellular fatty acid flux and glucose uptake in skeletal myotubes. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158678. [DOI: 10.1016/j.bbalip.2020.158678] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 02/25/2020] [Accepted: 02/28/2020] [Indexed: 12/17/2022]
|
28
|
Benitez-Amaro A, Revuelta-López E, Bornachea O, Cedó L, Vea À, Herrero L, Roglans N, Soler-Botija C, de Gonzalo-Calvo D, Nasarre L, Camino-López S, García E, Mato E, Blanco-Vaca F, Bayes-Genis A, Sebastian D, Laguna JC, Serra D, Zorzano A, Escola-Gil JC, Llorente-Cortes V. Low-density lipoprotein receptor-related protein 1 deficiency in cardiomyocytes reduces susceptibility to insulin resistance and obesity. Metabolism 2020; 106:154191. [PMID: 32112822 DOI: 10.1016/j.metabol.2020.154191] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 02/14/2020] [Accepted: 02/24/2020] [Indexed: 02/09/2023]
Abstract
BACKGROUND Low-density lipoprotein receptor-related protein 1 (LRP1) plays a key role in fatty acid metabolism and glucose homeostasis. In the context of dyslipemia, LRP1 is upregulated in the heart. Our aim was to evaluate the impact of cardiomyocyte LRP1 deficiency on high fat diet (HFD)-induced cardiac and metabolic alterations, and to explore the potential mechanisms involved. METHODS We used TnT-iCre transgenic mice with thoroughly tested suitability to delete genes exclusively in cardiomyocytes to generate an experimental mouse model with conditional Lrp1 deficiency in cardiomyocytes (TNT-iCre+-LRP1flox/flox). FINDINGS Mice with Lrp1-deficient cardiomyocytes (cm-Lrp1-/-) have a normal cardiac function combined with a favorable metabolic phenotype against HFD-induced glucose intolerance and obesity. Glucose intolerance protection was linked to higher hepatic fatty acid oxidation (FAO), lower liver steatosis and increased whole-body energy expenditure. Proteomic studies of the heart revealed decreased levels of cardiac pro-atrial natriuretic peptide (pro-ANP), which was parallel to higher ANP circulating levels. cm-Lrp1-/- mice showed ANP signaling activation that was linked to increased fatty acid (FA) uptake and increased AMPK/ ACC phosphorylation in the liver. Natriuretic peptide receptor A (NPR-A) antagonist completely abolished ANP signaling and metabolic protection in cm-Lrp1-/- mice. CONCLUSIONS These results indicate that an ANP-dependent axis controlled by cardiac LRP1 levels modulates AMPK activity in the liver, energy homeostasis and whole-body metabolism.
Collapse
Affiliation(s)
- Aleyda Benitez-Amaro
- Institute of Biochemical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain; Lipids and Cardiovascular Pathology Group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Elena Revuelta-López
- Heart Failure and Cardiac Regeneration (ICREC) Research Program, Health Sciences Research Institute Germans Trias i Pujol (IGTP), Badalona, Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Olga Bornachea
- Institute of Biochemical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain; Lipids and Cardiovascular Pathology Group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Lídia Cedó
- Metabolic Basis of Cardiovascular Risk, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Àngela Vea
- Institute of Biochemical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain
| | - Laura Herrero
- Department of Biochemistry and Physiology, School of Pharmacy, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Nuria Roglans
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Spain
| | - Carolina Soler-Botija
- Heart Failure and Cardiac Regeneration (ICREC) Research Program, Health Sciences Research Institute Germans Trias i Pujol (IGTP), Badalona, Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - David de Gonzalo-Calvo
- Institute of Biochemical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain; Lipids and Cardiovascular Pathology Group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Nasarre
- Institute of Biochemical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain
| | - Sandra Camino-López
- Institute of Biochemical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain
| | - Eduardo García
- Institute of Biochemical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain; Lipids and Cardiovascular Pathology Group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Eugenia Mato
- CIBER Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco Blanco-Vaca
- Metabolic Basis of Cardiovascular Risk, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Departament de Bioquímica, Biología Molecular i Biomedicina, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Antoni Bayes-Genis
- Heart Failure and Cardiac Regeneration (ICREC) Research Program, Health Sciences Research Institute Germans Trias i Pujol (IGTP), Badalona, Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; Cardiology Service and Heart Failure Unit, Hospital Universitari Germans Trias i Pujol, Badalona, Spain, Department of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - David Sebastian
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Joan Carles Laguna
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Spain
| | - Dolors Serra
- Department of Biochemistry and Physiology, School of Pharmacy, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Zorzano
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Joan Carles Escola-Gil
- Metabolic Basis of Cardiovascular Risk, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain.
| | - Vicenta Llorente-Cortes
- Institute of Biochemical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain; Lipids and Cardiovascular Pathology Group, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
29
|
Najt CP, Khan SA, Heden TD, Witthuhn BA, Perez M, Heier JL, Mead LE, Franklin MP, Karanja KK, Graham MJ, Mashek MT, Bernlohr DA, Parker L, Chow LS, Mashek DG. Lipid Droplet-Derived Monounsaturated Fatty Acids Traffic via PLIN5 to Allosterically Activate SIRT1. Mol Cell 2020; 77:810-824.e8. [PMID: 31901447 PMCID: PMC7036014 DOI: 10.1016/j.molcel.2019.12.003] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 10/17/2019] [Accepted: 12/03/2019] [Indexed: 12/20/2022]
Abstract
Lipid droplets (LDs) provide a reservoir for triacylglycerol storage and are a central hub for fatty acid trafficking and signaling in cells. Lipolysis promotes mitochondrial biogenesis and oxidative metabolism via a SIRT1/PGC-1α/PPARα-dependent pathway through an unknown mechanism. Herein, we identify that monounsaturated fatty acids (MUFAs) allosterically activate SIRT1 toward select peptide-substrates such as PGC-1α. MUFAs enhance PGC-1α/PPARα signaling and promote oxidative metabolism in cells and animal models in a SIRT1-dependent manner. Moreover, we characterize the LD protein perilipin 5 (PLIN5), which is known to enhance mitochondrial biogenesis and function, to be a fatty-acid-binding protein that preferentially binds LD-derived monounsaturated fatty acids and traffics them to the nucleus following cAMP/PKA-mediated lipolytic stimulation. Thus, these studies identify the first-known endogenous allosteric modulators of SIRT1 and characterize a LD-nuclear signaling axis that underlies the known metabolic benefits of MUFAs and PLIN5.
Collapse
Affiliation(s)
- Charles P Najt
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Salmaan A Khan
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Timothy D Heden
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Bruce A Witthuhn
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Minervo Perez
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Jason L Heier
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Linnea E Mead
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Mallory P Franklin
- Department of Food Science and Nutrition, University of Minnesota, Minneapolis, MN, USA
| | - Kenneth K Karanja
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | | | - Mara T Mashek
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Laurie Parker
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Lisa S Chow
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
| | - Douglas G Mashek
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA; Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
30
|
Roelands J, Garand M, Hinchcliff E, Ma Y, Shah P, Toufiq M, Alfaki M, Hendrickx W, Boughorbel S, Rinchai D, Jazaeri A, Bedognetti D, Chaussabel D. Long-Chain Acyl-CoA Synthetase 1 Role in Sepsis and Immunity: Perspectives From a Parallel Review of Public Transcriptome Datasets and of the Literature. Front Immunol 2019; 10:2410. [PMID: 31681299 PMCID: PMC6813721 DOI: 10.3389/fimmu.2019.02410] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 09/26/2019] [Indexed: 12/21/2022] Open
Abstract
A potential role for the long-chain acyl-CoA synthetase family member 1 (ACSL1) in the immunobiology of sepsis was explored during a hands-on training workshop. Participants first assessed the robustness of the potential gap in biomedical knowledge identified via an initial screen of public transcriptome data and of the literature associated with ACSL1. Increase in ACSL1 transcript abundance during sepsis was confirmed in several independent datasets. Querying the ACSL1 literature also confirmed the absence of reports associating ACSL1 with sepsis. Inferences drawn from both the literature (via indirect associations) and public transcriptome data (via correlation) point to the likely participation of ACSL1 and ACSL4, another family member, in inflammasome activation in neutrophils during sepsis. Furthermore, available clinical data indicate that levels of ACSL1 and ACSL4 induction was significantly higher in fatal cases of sepsis. This denotes potential translational relevance and is consistent with involvement in pathways driving potentially deleterious systemic inflammation. Finally, while ACSL1 expression was induced in blood in vitro by a wide range of pathogen-derived factors as well as TNF, induction of ACSL4 appeared restricted to flagellated bacteria and pathogen-derived TLR5 agonists and IFNG. Taken together, this joint review of public literature and omics data records points to two members of the acyl-CoA synthetase family potentially playing a role in inflammasome activation in neutrophils. Translational relevance of these observations in the context of sepsis and other inflammatory conditions remain to be investigated.
Collapse
Affiliation(s)
- Jessica Roelands
- Sidra Medicine, Doha, Qatar.,Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| | | | - Emily Hinchcliff
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ying Ma
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Parin Shah
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | | | | | | | | | - Amir Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | | |
Collapse
|
31
|
Lv Y, Cao Y, Gao Y, Yun J, Yu Y, Zhang L, Hu Z, Liu L, Xue J, Zhang G. Effect of ACSL3 Expression Levels on Preadipocyte Differentiation in Chinese Red Steppe Cattle. DNA Cell Biol 2019; 38:945-954. [DOI: 10.1089/dna.2018.4443] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Yang Lv
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yang Cao
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
- Key Laboratory of Beef Cattle Genetics and Breeding in Ministry of Agriculture and Rural Agriculture, Changchun, China
| | - Yi Gao
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
| | - Jinyan Yun
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
| | - Yongsheng Yu
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
| | - Lichun Zhang
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
| | - Zhongchang Hu
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
| | - Lixiang Liu
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Jiajia Xue
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Guoliang Zhang
- Branch of Animal Husbandry, Jilin Academy of Agricultural Science, Gongzhuling, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Laboratory of Beef Cattle Genetics and Breeding in Ministry of Agriculture and Rural Agriculture, Changchun, China
| |
Collapse
|
32
|
Adaptive evolution of the ACSL gene family in Carnivora. Genetica 2019; 147:141-148. [DOI: 10.1007/s10709-019-00057-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 03/06/2019] [Indexed: 10/27/2022]
|
33
|
Leung DTH, Rainczuk A, Nguyen T, Stephens A, Silke J, Fuller PJ, Chu S. Targeting XIAP and PPARγ in Granulosa Cell Tumors Alters Metabolic Signaling. J Proteome Res 2019; 18:1691-1702. [DOI: 10.1021/acs.jproteome.8b00917] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Dilys T. H. Leung
- Department of Molecular and Translational Science, Hudson Institute of Medical Research and the Monash University, Clayton, Victoria 3168, Australia
| | - Adam Rainczuk
- Department of Molecular and Translational Science, Hudson Institute of Medical Research and the Monash University, Clayton, Victoria 3168, Australia
| | - Trang Nguyen
- Department of Molecular and Translational Science, Hudson Institute of Medical Research and the Monash University, Clayton, Victoria 3168, Australia
| | - Andrew Stephens
- Department of Molecular and Translational Science, Hudson Institute of Medical Research and the Monash University, Clayton, Victoria 3168, Australia
| | - John Silke
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Peter J. Fuller
- Department of Molecular and Translational Science, Hudson Institute of Medical Research and the Monash University, Clayton, Victoria 3168, Australia
| | - Simon Chu
- Department of Molecular and Translational Science, Hudson Institute of Medical Research and the Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
34
|
Lee JY, Kim AR, Jung YH, Bu SY. Dissociation of Systemic Glucose Homeostasis from Triacylglyceride Accumulation by Reduced Acsl6 Expression in Skeletal Muscle. BIOTECHNOL BIOPROC E 2018. [DOI: 10.1007/s12257-018-0261-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
35
|
Signature molecules expressed differentially in a liver disease stage-specific manner by HIV-1 and HCV co-infection. PLoS One 2018; 13:e0202524. [PMID: 30138348 PMCID: PMC6107166 DOI: 10.1371/journal.pone.0202524] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 08/03/2018] [Indexed: 12/20/2022] Open
Abstract
To elucidate HIV-1 co-infection-induced acceleration of HCV liver disease and identify stage-specific molecular signatures, we applied a new high-resolution molecular screen, the Affymetrix GeneChip Human Transcriptome Array (HTA2.0), to HCV-mono- and HIV/HCV-co-infected liver specimens from subjects with early and advanced disease. Out of 67,528 well-annotated genes, we have analyzed the functional and statistical significance of 75 and 28 genes expressed differentially between early and advanced stages of HCV mono- and HIV/HCV co-infected patient liver samples, respectively. We also evaluated the expression of 25 and 17 genes between early stages of mono- and co-infected liver tissues and between advanced stages of mono- and co-infected patient's samples, respectively. Based on our analysis of fold-change in gene expression as a function of disease stage (i.e., early vs. advanced), coupled with consideration of the known relevant functions of these genes, we focused on four candidate genes, ACSL4, GNMT, IFI27, and miR122, which are expressed stage-specifically in HCV mono- and HIV-1/HCV co-infective liver disease and are known to play a pivotal role in regulating HCV-mediated hepatocellular carcinoma (HCC). Our qRT-PCR analysis of the four genes in patient liver specimens supported the microarray data. Protein products of each gene were detected in the endoplasmic reticulum (ER) where HCV replication takes place, and the genes' expression significantly altered replicability of HCV in the subgenomic replicon harboring regulatory genes of the JFH1 strain of HCV in Huh7.5.1. With respect to three well-known transferrable HIV-1 viral elements-Env, Nef, and Tat-Nef uniquely augmented replicon expression, while Tat, but not the others, substantially modulated expression of the candidate genes in hepatocytic cells. Combinatorial expression of these cellular and viral genes in the replicon cells further altered replicon expression. Taken together, these results showed that HIV-1 viral proteins can exacerbate liver pathology in the co-infected patients by disparate molecular mechanisms-directly or indirectly dysregulating HCV replication, even if lack of association of HCV load and end-stage liver disease in hemophilic patients were reported, and modulating expression of hepatocellular genes critical for disease progression. These findings also provide major insights into development of stage-specific hepatocellular biomarkers for improved diagnosis and prognosis of HCV-mediated liver disease.
Collapse
|
36
|
Expression Analysis of ACSL5 and Wnt2B in Human Congenital Pulmonary Airway Malformations. J Surg Res 2018; 232:128-136. [PMID: 30463708 DOI: 10.1016/j.jss.2018.06.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/22/2018] [Accepted: 06/06/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND The objective of this study was to determine acyl-CoA synthetase 5 (ACSL5) and Wnt2B expression patterns in human congenital pulmonary airway malformations (CPAMs) and to identify the possible roles of ACSL5 and Wnt2B in the pathogenesis of CPAM. METHODS Expression of ACSL5 and Wnt2B was evaluated by immunohistochemical staining, Western blotting, and quantitative real-time polymerase chain reaction, which were performed on surgical specimens of CPAM and adjacent normal lung tissues as controls. RESULTS Immunohistochemistry revealed that ACSL5 and Wnt2B immunopositive cells were predominantly detected in the mesenchymal cell nucleus, and there were lower expressions of ACSL5 and Wnt2B immunopositive cells in CPAM tissues than those in adjacent normal lung tissues. Western blotting and quantitative real-time polymerase chain reaction showed that ACSL5 and Wnt2B protein and mRNA expressions were significantly decreased in CPAM tissues as compared to the adjacent normal lung tissues (P < 0.05). In addition, there was a reduced level of ACSL5 relative to that of Wnt2B. CONCLUSIONS The decreased ACSL5 and Wnt2B expressions correlated with aberrations in pulmonary development and in the pathogenesis of CPAM, so downregulation of ACSL5 and Wnt2B could play an important role in the development of bronchial-alveolar structures in CPAM.
Collapse
|
37
|
Cai Y, Li H, Liu M, Pei Y, Zheng J, Zhou J, Luo X, Huang W, Ma L, Yang Q, Guo S, Xiao X, Li Q, Zeng T, Meng F, Francis H, Glaser S, Chen L, Huo Y, Alpini G, Wu C. Disruption of adenosine 2A receptor exacerbates NAFLD through increasing inflammatory responses and SREBP1c activity. Hepatology 2018; 68:48-61. [PMID: 29315766 PMCID: PMC6033664 DOI: 10.1002/hep.29777] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 12/15/2017] [Accepted: 12/29/2018] [Indexed: 01/04/2023]
Abstract
UNLABELLED Adenosine 2A receptor (A2A R) exerts protective roles in endotoxin- and/or ischemia-induced tissue damage. However, the role for A2A R in nonalcoholic fatty liver disease (NAFLD) remains largely unknown. We sought to examine the effects of global and/or myeloid cell-specific A2A R disruption on the aspects of obesity-associated NAFLD and to elucidate the underlying mechanisms. Global and/or myeloid cell-specific A2A R-disrupted mice and control mice were fed a high-fat diet (HFD) to induce NAFLD. In addition, bone marrow-derived macrophages and primary mouse hepatocytes were examined for inflammatory and metabolic responses. Upon feeding an HFD, both global A2A R-disrupted mice and myeloid cell-specific A2A R-defcient mice revealed increased severity of HFD-induced hepatic steatosis and inflammation compared with their respective control mice. In in vitro experiments, A2A R-deficient macrophages exhibited increased proinflammatory responses, and enhanced fat deposition of wild-type primary hepatocytes in macrophage-hepatocyte cocultures. In primary hepatocytes, A2A R deficiency increased the proinflammatory responses and enhanced the effect of palmitate on stimulating fat deposition. Moreover, A2A R deficiency significantly increased the abundance of sterol regulatory element-binding protein 1c (SREBP1c) in livers of fasted mice and in hepatocytes upon nutrient deprivation. In the absence of A2A R, SREBP1c transcription activity was significantly increased in mouse hepatocytes. CONCLUSION Taken together, our results demonstrate that disruption of A2A R in both macrophage and hepatocytes accounts for increased severity of NAFLD, likely through increasing inflammation and through elevating lipogenic events due to stimulation of SREBP1c expression and transcription activity. (Hepatology 2018;68:48-61).
Collapse
Affiliation(s)
- Yuli Cai
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA,Department of Endocrinology, Renmin Hospital, Wuhan University, Wuhan, Hubei 430060, China
| | - Honggui Li
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Mengyang Liu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Ya Pei
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Juan Zheng
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA,Department of Endocrinology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jing Zhou
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Xianjun Luo
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Wenya Huang
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Linqiang Ma
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA,Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China,Laboratory of Lipid & Glucose Metabolism, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qiuhua Yang
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA,Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Shaodong Guo
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Xiaoqiu Xiao
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China,Laboratory of Lipid & Glucose Metabolism, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qifu Li
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Tianshu Zeng
- Department of Endocrinology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Fanyin Meng
- Research, Central Texas Veterans Health Care System, Texas A&M University College of Medicine, Temple, TX 76504, USA,Department of Medical Physiology, Texas A&M University College of Medicine, Temple, TX 76504, USA
| | - Heather Francis
- Research, Central Texas Veterans Health Care System, Texas A&M University College of Medicine, Temple, TX 76504, USA,Department of Medical Physiology, Texas A&M University College of Medicine, Temple, TX 76504, USA
| | - Shannon Glaser
- Research, Central Texas Veterans Health Care System, Texas A&M University College of Medicine, Temple, TX 76504, USA,Department of Medical Physiology, Texas A&M University College of Medicine, Temple, TX 76504, USA
| | - Lulu Chen
- Department of Endocrinology, Union Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Yuqing Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA,Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Gianfranco Alpini
- Research, Central Texas Veterans Health Care System, Texas A&M University College of Medicine, Temple, TX 76504, USA,Department of Medical Physiology, Texas A&M University College of Medicine, Temple, TX 76504, USA,Contact information: Chaodong Wu, MD, PhD, College Station, TX 77843, Fax: 979 458 3129, ; or Gianfranco Alpini, PhD, Temple, TX 76504, ; Tel: 254 743 1041
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA,Contact information: Chaodong Wu, MD, PhD, College Station, TX 77843, Fax: 979 458 3129, ; or Gianfranco Alpini, PhD, Temple, TX 76504, ; Tel: 254 743 1041
| |
Collapse
|
38
|
Radif Y, Ndiaye H, Kalantzi V, Jacobs R, Hall A, Minogue S, Waugh MG. The endogenous subcellular localisations of the long chain fatty acid-activating enzymes ACSL3 and ACSL4 in sarcoma and breast cancer cells. Mol Cell Biochem 2018; 448:275-286. [PMID: 29450800 PMCID: PMC6182735 DOI: 10.1007/s11010-018-3332-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 02/09/2018] [Indexed: 12/22/2022]
Abstract
Fatty acid uptake and metabolism are often dysregulated in cancer cells. Fatty acid activation is a critical step that allows these biomolecules to enter cellular metabolic pathways such as mitochondrial β-oxidation for ATP generation or the lipogenic routes that generate bioactive lipids such as the inositol phospholipids. Fatty acid activation by the addition of coenzyme A is catalysed by a family of enzymes called the acyl CoA synthetase ligases (ACSL). Furthermore, enhanced expression of particular ACSL isoforms, such as ACSL4, is a feature of some more aggressive cancers and may contribute to the oncogenic phenotype. This study focuses on ACSL3 and ACSL4, closely related structural homologues that preferentially activate palmitate and arachidonate fatty acids, respectively. In this study, immunohistochemical screening of multiple soft tissue tumour arrays revealed that ACSL3 and ACSL4 were highly, but differentially, expressed in a subset of leiomyosarcomas, fibrosarcomas and rhabdomyosarcomas, with consistent cytoplasmic and granular stainings of tumour cells. The intracellular localisations of endogenously expressed ACSL3 and ACSL4 were further investigated by detailed subcellular fractionation analyses of HT1080 fibrosarcoma and MCF-7 breast cancer cells. ACSL3 distribution closely overlapped with proteins involved in trafficking from the trans-Golgi network and endosomes. In contrast, the ACSL4 localisation pattern more closely followed that of calnexin which is an endoplasmic reticulum resident chaperone. Confocal immunofluorescence imaging of MCF-7 cells confirmed the intracellular localisations of both enzymes. These observations reveal new information regarding the compartmentation of fatty acid metabolism in cancer cells.
Collapse
Affiliation(s)
- Yassmeen Radif
- Lipid & Membrane Biology Group, University College London, Floor U3, Royal Free Hospital Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Haarith Ndiaye
- Lipid & Membrane Biology Group, University College London, Floor U3, Royal Free Hospital Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Vasiliki Kalantzi
- Lipid & Membrane Biology Group, University College London, Floor U3, Royal Free Hospital Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Ruth Jacobs
- Lipid & Membrane Biology Group, University College London, Floor U3, Royal Free Hospital Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Andrew Hall
- Sheila Sherlock Liver Centre, Royal Free London NHS Foundation Trust and UCL Institute for Liver and Digestive Health, University College London, London, UK
| | - Shane Minogue
- Lipid & Membrane Biology Group, University College London, Floor U3, Royal Free Hospital Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - Mark G Waugh
- Lipid & Membrane Biology Group, University College London, Floor U3, Royal Free Hospital Campus, Rowland Hill Street, London, NW3 2PF, UK.
| |
Collapse
|
39
|
Abstract
Triglyceride molecules represent the major form of storage and transport of fatty acids within cells and in the plasma. The liver is the central organ for fatty acid metabolism. Fatty acids accrue in liver by hepatocellular uptake from the plasma and by de novo biosynthesis. Fatty acids are eliminated by oxidation within the cell or by secretion into the plasma within triglyceride-rich very low-density lipoproteins. Notwithstanding high fluxes through these pathways, under normal circumstances the liver stores only small amounts of fatty acids as triglycerides. In the setting of overnutrition and obesity, hepatic fatty acid metabolism is altered, commonly leading to the accumulation of triglycerides within hepatocytes, and to a clinical condition known as nonalcoholic fatty liver disease (NAFLD). In this review, we describe the current understanding of fatty acid and triglyceride metabolism in the liver and its regulation in health and disease, identifying potential directions for future research. Advances in understanding the molecular mechanisms underlying the hepatic fat accumulation are critical to the development of targeted therapies for NAFLD. © 2018 American Physiological Society. Compr Physiol 8:1-22, 2018.
Collapse
Affiliation(s)
- Michele Alves-Bezerra
- Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, USA
| | - David E Cohen
- Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, USA
| |
Collapse
|
40
|
Franklin MP, Sathyanarayan A, Mashek DG. Acyl-CoA Thioesterase 1 (ACOT1) Regulates PPARα to Couple Fatty Acid Flux With Oxidative Capacity During Fasting. Diabetes 2017; 66:2112-2123. [PMID: 28607105 PMCID: PMC5521868 DOI: 10.2337/db16-1519] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 05/17/2017] [Indexed: 12/18/2022]
Abstract
Hepatic acyl-CoA thioesterase 1 (ACOT1) catalyzes the conversion of acyl-CoAs to fatty acids (FAs) and CoA. We sought to determine the role of ACOT1 in hepatic lipid metabolism in C57Bl/6J male mice 1 week after adenovirus-mediated Acot1 knockdown. Acot1 knockdown reduced liver triglyceride (TG) as a result of enhanced TG hydrolysis and subsequent FA oxidation. In vitro experiments demonstrated that Acot1 knockdown led to greater TG turnover and FA oxidation, suggesting that ACOT1 is important for controlling the rate of FA oxidation. Despite increased FA oxidation, Acot1 knockdown reduced the expression of peroxisome proliferator-activated receptor α (PPARα) target genes, whereas overexpression increased PPARα reporter activity, suggesting ACOT1 regulates PPARα by producing FA ligands. Moreover, ACOT1 exhibited partial nuclear localization during fasting and cAMP/cAMP-dependent protein kinase signaling, suggesting local regulation of PPARα. As a consequence of increased FA oxidation and reduced PPARα activity, Acot1 knockdown enhanced hepatic oxidative stress and inflammation. The effects of Acot1 knockdown on PPARα activity, oxidative stress, and inflammation were rescued by supplementation with Wy-14643, a synthetic PPARα ligand. We demonstrate through these results that ACOT1 regulates fasting hepatic FA metabolism by balancing oxidative flux and capacity.
Collapse
Affiliation(s)
- Mallory P Franklin
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, MN
| | | | - Douglas G Mashek
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, MN
| |
Collapse
|
41
|
Guo X, Shu C, Li H, Pei Y, Woo SL, Zheng J, Liu M, Xu H, Botchlett R, Guo T, Cai Y, Gao X, Zhou J, Chen L, Li Q, Xiao X, Xie L, Zhang KK, Ji JY, Huo Y, Meng F, Alpini G, Li P, Wu C. Cyclic GMP-AMP Ameliorates Diet-induced Metabolic Dysregulation and Regulates Proinflammatory Responses Distinctly from STING Activation. Sci Rep 2017; 7:6355. [PMID: 28743914 PMCID: PMC5526935 DOI: 10.1038/s41598-017-05884-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/26/2017] [Indexed: 01/22/2023] Open
Abstract
Endogenous cyclic GMP-AMP (cGAMP) binds and activates STING to induce type I interferons. However, whether cGAMP plays any roles in regulating metabolic homeostasis remains unknown. Here we show that exogenous cGAMP ameliorates obesity-associated metabolic dysregulation and uniquely alters proinflammatory responses. In obese mice, treatment with cGAMP significantly decreases diet-induced proinflammatory responses in liver and adipose tissues and ameliorates metabolic dysregulation. Strikingly, cGAMP exerts cell-type-specific anti-inflammatory effects on macrophages, hepatocytes, and adipocytes, which is distinct from the effect of STING activation by DMXAA on enhancing proinflammatory responses. While enhancing insulin-stimulated Akt phosphorylation in hepatocytes and adipocytes, cGAMP weakens the effects of glucagon on stimulating hepatocyte gluconeogenic enzyme expression and glucose output and blunts palmitate-induced hepatocyte fat deposition in an Akt-dependent manner. Taken together, these results suggest an essential role for cGAMP in linking innate immunity and metabolic homeostasis, indicating potential applications of cGAMP in treating obesity-associated inflammatory and metabolic diseases.
Collapse
Affiliation(s)
- Xin Guo
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Chang Shu
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, 77843, USA
| | - Honggui Li
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Ya Pei
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Shih-Lung Woo
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Juan Zheng
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Mengyang Liu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Hang Xu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Rachel Botchlett
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Ting Guo
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Yuli Cai
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Xinsheng Gao
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, College Station, Texas, 77843, USA
| | - Jing Zhou
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Lu Chen
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Qifu Li
- Department of Endocrinology and the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoqiu Xiao
- Department of Endocrinology and the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.,The Laboratory of Lipid & Glucose Metabolism, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Linglin Xie
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA
| | - Ke K Zhang
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58202, USA
| | - Jun-Yuan Ji
- Department of Endocrinology and the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yuqing Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.,Drug Discovery Center, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Fanyin Meng
- Departments of Medical Physiology and Medicine, Texas A&M University Health Science Center, Temple, TX, 76504, USA
| | - Gianfranco Alpini
- Departments of Medical Physiology and Medicine, Texas A&M University Health Science Center, Temple, TX, 76504, USA
| | - Pingwei Li
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, 77843, USA.
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
42
|
CDCP1 drives triple-negative breast cancer metastasis through reduction of lipid-droplet abundance and stimulation of fatty acid oxidation. Proc Natl Acad Sci U S A 2017; 114:E6556-E6565. [PMID: 28739932 DOI: 10.1073/pnas.1703791114] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is notoriously aggressive with high metastatic potential, which has recently been linked to high rates of fatty acid oxidation (FAO). Here we report the mechanism of lipid metabolism dysregulation in TNBC through the prometastatic protein, CUB-domain containing protein 1 (CDCP1). We show that a "low-lipid" phenotype is characteristic of breast cancer cells compared with normal breast epithelial cells and negatively correlates with invasiveness in 3D culture. Using coherent anti-Stokes Raman scattering and two-photon excited fluorescence microscopy, we show that CDCP1 depletes lipids from cytoplasmic lipid droplets (LDs) through reduced acyl-CoA production and increased lipid utilization in the mitochondria through FAO, fueling oxidative phosphorylation. These findings are supported by CDCP1's interaction with and inhibition of acyl CoA-synthetase ligase (ACSL) activity. Importantly, CDCP1 knockdown increases LD abundance and reduces TNBC 2D migration in vitro, which can be partially rescued by the ACSL inhibitor, Triacsin C. Furthermore, CDCP1 knockdown reduced 3D invasion, which can be rescued by ACSL3 co-knockdown. In vivo, inhibiting CDCP1 activity with an engineered blocking fragment (extracellular portion of cleaved CDCP1) lead to increased LD abundance in primary tumors, decreased metastasis, and increased ACSL activity in two animal models of TNBC. Finally, TNBC lung metastases have lower LD abundance than their corresponding primary tumors, indicating that LD abundance in primary tumor might serve as a prognostic marker for metastatic potential. Our studies have important implications for the development of TNBC therapeutics to specifically block CDCP1-driven FAO and oxidative phosphorylation, which contribute to TNBC migration and metastasis.
Collapse
|
43
|
Huang Y, Ye T, Liu C, Fang F, Chen Y, Dong Y. Maternal high-fat diet during pregnancy and lactation affects hepatic lipid metabolism in early life of offspring rat. J Biosci 2017; 42:311-319. [DOI: 10.1007/s12038-017-9675-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
44
|
TNF-α stimulates endothelial palmitic acid transcytosis and promotes insulin resistance. Sci Rep 2017; 7:44659. [PMID: 28304381 PMCID: PMC5356338 DOI: 10.1038/srep44659] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 02/13/2017] [Indexed: 01/15/2023] Open
Abstract
Persistent elevation of plasma TNF-α is a marker of low grade systemic inflammation. Palmitic acid (PA) is the most abundant type of saturated fatty acid in human body. PA is bound with albumin in plasma and could not pass through endothelial barrier freely. Albumin-bound PA has to be transported across monolayer endothelial cells through intracellular transcytosis, but not intercellular diffusion. In the present study, we discovered that TNF-α might stimulate PA transcytosis across cardiac microvascular endothelial cells, which further impaired the insulin-stimulated glucose uptake by cardiomyocytes and promoted insulin resistance. In this process, TNF-α-stimulated endothelial autophagy and NF-κB signaling crosstalk with each other and orchestrate the whole event, ultimately result in increased expression of fatty acid transporter protein 4 (FATP4) in endothelial cells and mediate the increased PA transcytosis across microvascular endothelial cells. Hopefully the present study discovered a novel missing link between low grade systemic inflammation and insulin resistance.
Collapse
|
45
|
Wooten JS, Nick TN, Seija A, Poole KE, Stout KB. High-Fructose Intake Impairs the Hepatic Hypolipidemic Effects of a High-Fat Fish-Oil Diet in C57BL/6 Mice. J Clin Exp Hepatol 2016; 6:265-274. [PMID: 28003715 PMCID: PMC5157917 DOI: 10.1016/j.jceh.2016.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 09/01/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Overnutrition of saturated fats and fructose is one of the major factors for the development of nonalcoholic fatty liver disease. Because omega-3 polyunsaturated fatty acids (n-3fa) have established lipid lowering properties, we tested the hypothesis that n-3fa prevents high-fat and fructose-induced fatty liver disease in mice. METHODS Male C57BL/6J mice were randomly assigned to one of the following diet groups for 14 weeks: normal diet (ND), high-fat lard-based diet (HFD), HFD with fructose (HFD + Fru), high-fat fish-oil diet (FOD), or FOD + Fru. RESULTS Despite for the development of obesity and insulin resistance, FOD had 65.3% lower (P < 0.001) hepatic triglyceride levels than HFD + Fru, which was blunted to a 38.5% difference (P = 0.173) in FOD + Fru. The lower hepatic triglyceride levels were associated with a lower expression of lipogenic genes LXRα and FASN, as well as the expression of genes associated with fatty acid uptake and triglyceride synthesis, CD36 and SCD1, respectively. Conversely, the blunted hypotriglyceride effect of FOD + Fru was associated with a higher expression of CD36 and SCD1. CONCLUSIONS During overnutrition, a diet rich in n-3fa may prevent the severity of hepatic steatosis; however, when juxtaposed with a diet high in fructose, the deleterious effects of overnutrition blunted the hypolipidemic effects of n-3fa.
Collapse
Key Words
- ACC1, acetyl-CoA carboxylase-1
- CPT1a, carnitine palmitoyltransferase 1a
- ChREBP, carbohydrate response element binding protein
- FASN, fatty acid synthase
- FFA, free fatty acid
- LPL, lipoprotein lipase
- LXRα, liver-X-receptor
- MTTP, microsomal triglyceride transfer protein
- NAFLD, nonalcoholic fatty liver disease
- PPARα, peroxisome proliferator activated receptor α
- PPARγ, peroxisome proliferator activated receptor γ
- SCD1, stearoyl-CoA desaturase 1
- SREBP1c, sterol response element binding protein
- T2DM, type 2 diabetes mellitus
- TRL, triglyceride-rich lipoproteins
- VLDL, very low-density lipoprotein
- fructose
- lipid metabolism
- lipotoxicity
- n-3fa, omega-3 polyunsaturated fatty acids
- omega-3 polyunsaturated fatty acids
- overnutrition
Collapse
Affiliation(s)
- Joshua S. Wooten
- Address for correspondence: Joshua S. Wooten, Department of Applied Health, Southern Illinois University Edwardsville, Campus Box 1126, Edwardsville, IL 62026-1126, United States. Fax: +1 618 650 3719.Department of Applied Health, Southern Illinois University EdwardsvilleCampus Box 1126EdwardsvilleIL62026-1126United States
| | | | | | | | | |
Collapse
|
46
|
Teodoro BG, Sampaio IH, Bomfim LHM, Queiroz AL, Silveira LR, Souza AO, Fernandes AMAP, Eberlin MN, Huang TY, Zheng D, Neufer PD, Cortright RN, Alberici LC. Long-chain acyl-CoA synthetase 6 regulates lipid synthesis and mitochondrial oxidative capacity in human and rat skeletal muscle. J Physiol 2016; 595:677-693. [PMID: 27647415 DOI: 10.1113/jp272962] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/11/2016] [Indexed: 12/29/2022] Open
Abstract
KEY POINTS Long-chain acyl-CoA synthetase 6 (ACSL6) mRNA is present in human and rat skeletal muscle, and is modulated by nutritional status: exercise and fasting decrease ACSL6 mRNA, whereas acute lipid ingestion increase its expression. ACSL6 genic inhibition in rat primary myotubes decreased lipid accumulation, as well as activated the higher mitochondrial oxidative capacity programme and fatty acid oxidation through the AMPK/PGC1-α pathway. ACSL6 overexpression in human primary myotubes increased phospholipid species and decreased oxidative metabolism. ABSTRACT Long-chain acyl-CoA synthetases (ACSL 1 to 6) are key enzymes regulating the partitioning of acyl-CoA species toward different metabolic fates such as lipid synthesis or β-oxidation. Despite our understanding of ecotopic lipid accumulation in skeletal muscle being associated with metabolic diseases such as obesity and type II diabetes, the role of specific ACSL isoforms in lipid synthesis remains unclear. In the present study, we describe for the first time the presence of ACSL6 mRNA in human skeletal muscle and the role that ACSL6 plays in lipid synthesis in both rodent and human skeletal muscle. ACSL6 mRNA was observed to be up-regulated by acute high-fat meal ingestion in both rodents and humans. In rats, we also demonstrated that fasting and chronic aerobic training negatively modulated the ACSL6 mRNA and other genes of lipid synthesis. Similar results were obtained following ACSL6 knockdown in rat myotubes, which was associated with a decreased accumulation of TAGs and lipid droplets. Under the same knockdown condition, we further demonstrate an increase in fatty acid content, p-AMPK, mitochondrial content, mitochondrial respiratory rates and palmitate oxidation. These results were associated with increased PGC-1α, UCP2 and UCP3 mRNA and decreased reactive oxygen species production. In human myotubes, ACSL6 overexpression reduced palmitate oxidation and PGC-1α mRNA. In conclusion, ACSL6 drives acyl-CoA toward lipid synthesis and its downregulation improves mitochondrial biogenesis, respiratory capacity and lipid oxidation. These outcomes are associated with the activation of the AMPK/PGC1-α pathway.
Collapse
Affiliation(s)
- Bruno G Teodoro
- Department of Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil.,Federal Institute of Education Science and Technology of São Paulo, Sertãozinho, São Paulo, Brazil
| | - Igor H Sampaio
- Department of Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Lucas H M Bomfim
- Department of Structural and Functional Biology, Institute of Biology
| | - André L Queiroz
- Department of Structural and Functional Biology, Institute of Biology
| | | | - Anderson O Souza
- Department of Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Anna M A P Fernandes
- ThoMSon Mass Spectrometry Laboratory, Institute of Chemistry, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Marcos N Eberlin
- ThoMSon Mass Spectrometry Laboratory, Institute of Chemistry, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | | | - Donghai Zheng
- Department of Kinesiology.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| | - P Darrell Neufer
- Department of Physiology.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| | - Ronald N Cortright
- Department of Kinesiology.,Department of Physiology.,East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, USA
| | - Luciane C Alberici
- Department of Physics and Chemistry, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
47
|
Genomics of human fatty liver disease reveal mechanistically linked lipid droplet-associated gene regulations in bland steatosis and nonalcoholic steatohepatitis. Transl Res 2016; 177:41-69. [PMID: 27376874 DOI: 10.1016/j.trsl.2016.06.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 05/13/2016] [Accepted: 06/08/2016] [Indexed: 12/11/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common disorder hallmarked by excessive lipid deposits. Based on our recent research on lipid droplet (LD) formation in hepatocytes, we investigated LD-associated gene regulations in NAFLD of different grades, that is, steatosis vs steatohepatitis by comparing liver biopsies from healthy controls (N = 13) and NAFLD patients (N = 102). On average, more than 700 differentially expressed genes (DEGs) were identified of which 146 are mechanistically linked to LD formation. We identified 51 LD-associated DEGs frequently regulated in patient samples (range ≥5 to ≤102) with the liver-receptor homolog-1(NR5A2), that is, a key regulator of cholesterol metabolism being commonly repressed among 100 patients examined. With bland steatosis, notable regulations involved hypoxia-inducible lipid droplet-associated-protein and diacylglycerol-O-acyltransferase-2 renowned for their role in LD-growth. Conversely, nonalcoholic steatohepatitis-associated DEGs coded for epidermal growth factor receptor and TLR4 signaling with decreased expression of the GTPase Rab5 and the lipid phosphohydrolase PPAP2B thus highlighting adaptive responses to inflammation, LDL-mediated endocytosis and lipogenesis, respectively. Studies with steatotic primary human hepatocyte cultures demonstrated induction of LD-associated PLIN2, CIDEC, DNAAF1, whereas repressed expression of CPT1A, ANGPTL4, and PKLR informed on burdened mitochondrial metabolism. Equally, repressed expression of the B-lymphocyte chemoattractant CXCL13 and STAT4 as well as induced FGF21 evidenced amelioration of steatosis-related inflammation. In-vitro/in-vivo patient sample comparisons confirmed C-reactive protein, SOCS3, NR5A2, and SOD2 as commonly regulated. Lastly, STRING network analysis highlighted potential "druggable" targets with PLIN2, CIDEC, and hypoxia-inducible lipid droplet-associated-protein being confirmed by immunofluorescence microscopy. In conclusion, steatosis and steatohepatitis specific gene regulations informed on the pathogenesis of NAFLD to broaden the perspective of targeted therapies.
Collapse
|
48
|
Huang Y, Huang S, Lam SM, Liu Z, Shui G, Zhang YQ. Acsl, the Drosophila ortholog of intellectual-disability-related ACSL4, inhibits synaptic growth by altered lipids. J Cell Sci 2016; 129:4034-4045. [PMID: 27656110 DOI: 10.1242/jcs.195032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/16/2016] [Indexed: 12/17/2022] Open
Abstract
Nervous system development and function are tightly regulated by metabolic processes, including the metabolism of lipids such as fatty acids. Mutations in long-chain acyl-CoA synthetase 4 (ACSL4) are associated with non-syndromic intellectual disabilities. We previously reported that Acsl, the Drosophila ortholog of mammalian ACSL3 and ACSL4, inhibits neuromuscular synapse growth by suppressing bone morphogenetic protein (BMP) signaling. Here, we report that Acsl regulates the composition of fatty acids and membrane lipids, which in turn affects neuromuscular junction (NMJ) synapse development. Acsl mutant brains had a decreased abundance of C16:1 fatty acyls; restoration of Acsl expression abrogated NMJ overgrowth and the increase in BMP signaling. A lipidomic analysis revealed that Acsl suppressed the levels of three lipid raft components in the brain, including mannosyl glucosylceramide (MacCer), phosphoethanolamine ceramide and ergosterol. The MacCer level was elevated in Acsl mutant NMJs and, along with sterol, promoted NMJ overgrowth, but was not associated with the increase in BMP signaling in the mutants. These findings suggest that Acsl inhibits NMJ growth by stimulating C16:1 fatty acyl production and concomitantly suppressing raft-associated lipid levels.
Collapse
Affiliation(s)
- Yan Huang
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Sheng Huang
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.,Sino-Danish College, Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Sin Man Lam
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhihua Liu
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Guanghou Shui
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yong Q Zhang
- State Key Laboratory for Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
49
|
Targeting Oct1 genomic function inhibits androgen receptor signaling and castration-resistant prostate cancer growth. Oncogene 2016; 35:6350-6358. [PMID: 27270436 DOI: 10.1038/onc.2016.171] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 02/22/2016] [Accepted: 04/12/2016] [Indexed: 12/25/2022]
Abstract
Androgen receptor (AR) functions as a ligand-dependent transcription factor to regulate its downstream signaling for prostate cancer progression. AR complex formation by multiple transcription factors is important for enhancer activity and transcriptional regulation. However, the significance of such collaborative transcription factors has not been fully understood. In this study, we show that Oct1, an AR collaborative factor, coordinates genome-wide AR signaling for prostate cancer growth. Using global analysis by chromatin immunoprecipitation sequencing (ChIP-seq), we found that Oct1 is recruited to AR-binding enhancer/promoter regions and facilitates androgen signaling. Moreover, a major target of AR/Oct1 complex, acyl-CoA synthetase 3 (ACSL3), contributes to tumor growth in nude mice, and its high expression is associated with poor prognosis in prostate cancer patients. Next, we examined the therapeutic effects of pyrrole-imidazole polyamides that target the Oct1-binding sequence identified in the center of the ACSL3 AR-binding site. We observed that treatment with Oct1 polyamide severely blocked the Oct1 binding at the ACSL3 enhancer responsible for its transcriptional activity and ACSL3 induction. In addition, Oct1 polyamides suppressed castration-resistant tumor growth and specifically repressed global Oct1 chromatin association and androgen signaling in prostate cancer cells, with few nonspecific effects on basal promoter activity. Thus, targeting Oct1 binding could be a novel therapeutic strategy for AR-activated castration-resistant prostate cancer.
Collapse
|
50
|
Pinolenic Acid Downregulates Lipid Anabolic Pathway in HepG2 Cells. Lipids 2016; 51:847-55. [DOI: 10.1007/s11745-016-4149-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 03/31/2016] [Indexed: 12/15/2022]
|