1
|
Ma Z, Pan S, Yang Y, Ren H, Yin S, Chen Q, An Z, Zhao X, Xu Z. Lipid droplets: Emerging therapeutic targets for age-related metabolic diseases. Ageing Res Rev 2025; 108:102758. [PMID: 40300696 DOI: 10.1016/j.arr.2025.102758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/01/2025]
Abstract
Lipids metabolism is crucial in regulating aging and metabolic diseases. Lipid droplets (LDs) are dynamic, complex organelles responsible for the storage and release of neutral lipids, essential for maintaining lipid homeostasis and energy metabolism. Aging accelerates the accumulation of LDs, functional deterioration, and metabolic disorders, thereby inducing age-related metabolic diseases (ARMDs). This review examines published datasets on the association between LDs and ARMDs, focusing on the structure and function of LDs, their interactions with other organelles, and associated proteins. Furthermore, we explore the potential mechanisms by which LDs mediate the onset of ARMDs, including Alzheimer's disease (AD), sarcopenia, metabolic cardiomyopathy, non-alcoholic fatty liver disease (NAFLD), and cancer. Lastly, we discuss intervention strategies aimed at targeting LDs to improve outcomes in ARMDs, including exercise, dietary, and pharmacological interventions.
Collapse
Affiliation(s)
- Zheying Ma
- School of Physical Education and Health Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Shou Pan
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an 710119, China
| | - Yaming Yang
- School of Physical Education and Health Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Huiqian Ren
- School of Physical Education and Health Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Sikun Yin
- School of Physical Education and Health Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Qianyu Chen
- School of Physical Education and Health Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Zhenxian An
- School of Physical Education and Health Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China
| | - Xiaoqin Zhao
- School of Physical Education and Health Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China.
| | - Zujie Xu
- School of Physical Education and Health Engineering, Taiyuan University of Technology, Taiyuan, Shanxi 030024, China.
| |
Collapse
|
2
|
Li X, Han X, Yan H, Zhu H, Wang H, Li D, Tian Y, Su Y. From gut microbiota to host genes: A dual-regulatory pathway driving body weight variation in dagu chicken (Gallus gallus domesticus). Poult Sci 2025; 104:105067. [PMID: 40239312 PMCID: PMC12032334 DOI: 10.1016/j.psj.2025.105067] [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: 01/12/2025] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
During the growth and development of animals, there is an interaction between the gut microbiota and the host genotype. The host genotype can regulate the microbiota, and in turn, the microbiota can influence host gene expression, thereby affecting the animal's production performance. This study explored the dynamic interplay between the gut microbiota and host gene expression in body weight variation in Dagu chicken, an indigenous poultry genetic resource in China. We characterized mucosa-associated microbiota across four gastrointestinal segments (duodenum, jejunum, ileum, cecum) and ileocecal chyme microbiota in 12-week-old Dagu chickens stratified by divergent body weight phenotypes, while simultaneously quantifying region-specific intestinal epithelial transcriptional regulation. 16S rDNA sequencing was employed to identify Firmicutes as the predominant bacterial phylum, with notable differences in the abundance of specific genera (e.g., Ligilactobacillus and Lactobacillus) being observed between the high- or low-body-weight groups. Enhanced biosynthesis pathways were functionally predicted in heavier roosters, whereas reduced nutrient metabolism pathways were contrasted. A conserved functional concordance was observed between regionally predominant differential microbiota and the physiological specialization of corresponding intestinal niches. Functional analysis revealed that the high-body-weight group demonstrated superior capabilities in microbial biosynthesis, whereas the low-body-weight group exhibited enhanced microbial metabolic activity. NAA80 was identified as the common differentially expressed gene across all intestinal epithelial tissues. The Gene Ontology and KEGG pathway analyses revealed elevated nutrient absorption efficiency in the high-body-weight group, while the low-body-weight group demonstrated accelerated cellular renewal rates and shorter cycles. Correlation analysis identified significant associations between gut microbiota and host genes expression profiles, with the majority of correlations being positive. These results suggest a coordinated interaction between microbial communities and host genetic regulation, potentially driving phenotypic differences in body weight performance.
Collapse
Affiliation(s)
- Xiaohan Li
- College of Animal Husbandry and Veterinary, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China; Liaoning Provincial Key Laboratory of Animal Product Quality and Safety, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China
| | - Xueru Han
- College of Animal Husbandry and Veterinary, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China; Liaoning Provincial Key Laboratory of Animal Product Quality and Safety, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China
| | - Huan Yan
- College of Animal Husbandry and Veterinary, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China; Liaoning Provincial Key Laboratory of Animal Product Quality and Safety, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China
| | - Hongyan Zhu
- College of Basic Medical Science, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China
| | - Hongcai Wang
- College of Animal Husbandry and Veterinary, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China; Liaoning Provincial Key Laboratory of Animal Product Quality and Safety, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China
| | - Desheng Li
- College of Animal Husbandry and Veterinary, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China; Liaoning Provincial Key Laboratory of Animal Product Quality and Safety, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China
| | - Yumin Tian
- College of Animal Husbandry and Veterinary, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China; Liaoning Provincial Key Laboratory of Animal Product Quality and Safety, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China
| | - Yuhong Su
- College of Animal Husbandry and Veterinary, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China; Liaoning Provincial Key Laboratory of Animal Product Quality and Safety, Jinzhou Medical University, Jinzhou 121001, Liaoning, PR China.
| |
Collapse
|
3
|
Zuo R, Wang M, Wang YT, ShenTu Y, Moura AK, Zhou Y, Roudbari K, Hu JZ, Li PL, Hao J, Li X, Zhang Y. Ablation of Hepatic Asah1 Gene Disrupts Hepatic Lipid Homeostasis and Promotes Fibrotic Nonalcoholic Steatohepatitis in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:542-560. [PMID: 39719015 PMCID: PMC11983695 DOI: 10.1016/j.ajpath.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/14/2024] [Accepted: 11/06/2024] [Indexed: 12/26/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) encompasses a spectrum of chronic liver conditions, ranging from simple steatosis to nonalcoholic steatohepatitis, which may progress to fibrosis/cirrhosis. Here, the GSE163211 data set was analyzed, and Asah1 (encoding acid ceramidase) was identified as a crucial lysosomal gene that positively correlated with NAFLD stages in obese patients. To evaluate the role of Asah1 in the progression of NAFLD, Asah1fl/fl/Albcre mice (hepatocyte-specific deletion of Asah1) and Asah1 floxed (Asah1fl/fl/wild-type) mice were fed with either a normal diet or a high-fat, high-cholesterol paigen diet (PD) for 20 weeks. Hepatocyte-specific Asah1 ablation markedly aggravated PD-induced hepatic steatosis, hepatitis, and apoptosis, and resulted in marked fibrotic changes. In addition, Asah1 gene ablation exacerbated PD-induced portal venous hemodynamic abnormality. In cultured hepatocytes, Asah1 gene knockdown resulted in increased ceramide and cholesterol levels but did not affect triglyceride level. Knocking down Asah1 gene also exhibited broad impacts on lipid homeostasis pathways, including lipogenesis, fatty acid uptake, fatty acid oxidation, and lipid transport. Furthermore, Asah1 knockdown resulted in increased endoplasmic reticulum stress and lipid droplet biogenesis. Finally, Asah1 gene knockdown impaired chaperone-mediated autophagy. These results suggest that Asah1 functions as an important regulator of hepatic lipid homeostasis, and its deficiency exacerbates hepatocyte lipotoxicity and injury, and promotes the development of fibrotic nonalcoholic steatohepatitis.
Collapse
Affiliation(s)
- Rui Zuo
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Mi Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas; Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun-Ting Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - YangPing ShenTu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas; Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Alexandra K Moura
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Ying Zhou
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas; Department Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kiana Roudbari
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Jenny Z Hu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - JiuKuan Hao
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Xiang Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas.
| |
Collapse
|
4
|
Wu X, Miller JA, Lee BTK, Wang Y, Ruedl C. Reducing microglial lipid load enhances β amyloid phagocytosis in an Alzheimer's disease mouse model. SCIENCE ADVANCES 2025; 11:eadq6038. [PMID: 39908361 PMCID: PMC11797491 DOI: 10.1126/sciadv.adq6038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 01/03/2025] [Indexed: 02/07/2025]
Abstract
Macrophages accumulate lipid droplets (LDs) under stress and inflammatory conditions. Despite the presence of LD-loaded macrophages in many tissues, including the brain, their contribution to neurodegenerative disorders remains elusive. This study investigated the role of lipid metabolism in Alzheimer's disease (AD) by assessing the contribution of LD-loaded brain macrophages, including microglia and border-associated macrophages (BAMs), in an AD mouse model. Particularly, BAMs and activated CD11c+ microglia localized near β amyloid (Aβ) plaques exhibited a pronounced lipid-associated gene signature and a high LD load. Having observed that elevated intracellular LD content correlated inversely with microglial phagocytic activities, we subsequently inhibited LD formation specifically in CX3CR1+ brain macrophages using an inducible APP-KI/Fit2iΔMφ transgenic mouse model. We demonstrated that reducing LD content in microglia and CX3CR1+ BAMs remarkably improved their phagocytic ability. Furthermore, lowering microglial LDs consistently enhanced their efferocytosis capacities and notably reduced Aβ deposition in the brain parenchyma. Therefore, mitigating LD accumulation in brain macrophages provides perspectives for AD treatment.
Collapse
Affiliation(s)
- Xiaoting Wu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - James Alastair Miller
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Bernett Teck Kwong Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Yulan Wang
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Christiane Ruedl
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
5
|
Salazar-Villacorta A, Bond LM, Kim L, Anagnostopoulou K, Scardamaglia A, Filippakopoulou E, Ververi A, Efthymiou S, Dinopoulos A, Murphy D, Karadima G, Koutsis G, Kaliakatsos M, Houlden H, Walther TC, Farese RV. Partial loss of FITM2 function causes hereditary spastic paraplegia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.23.24319660. [PMID: 39974099 PMCID: PMC11838939 DOI: 10.1101/2025.01.23.24319660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
FITM2 encodes fat-storage inducing transmembrane protein 2 (FIT2), a lipid diphosphatase in the ER that cleaves acyl-CoAs and is crucial for ER homeostasis. In humans, homozygous null mutations in FITM2 are associated with a syndrome characterized by deafness and dystonia. Here, we report two families with hereditary spastic paraplegia (HSP) in whom exome sequencing revealed compound heterozygosity for FITM2 mutations. In each family, the affected probands carry one putative null allele and one G100R missense allele. Functional analyses demonstrated that the G100R allele is hypomorphic, with FIT2 protein levels reduced to 20% of wild type, leading to proportionately decreased enzyme activity. The occurrence of similar HSP disease phenotypes and the same hypomorphic mutation in these families suggests that the G100R mutation and its associated reduced enzyme activity represent a newly recognized clinical manifestation of FITM2 mutations, expanding the spectrum of conditions associated with this gene.
Collapse
|
6
|
Calvo I, Fresnedo O, Mosteiro L, López JI, Larrinaga G, Fernández JA. Lipid imaging mass spectrometry: Towards a new molecular histology. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159568. [PMID: 39369885 DOI: 10.1016/j.bbalip.2024.159568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/25/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Lipid research is attracting greater attention, as these molecules are key components to understand cell metabolism and the connection between genotype and phenotype. The study of lipids has also been fueled by the development of new and powerful technologies, able to identify an increasing number of species in a single run and at decreasing concentrations. One of such key developments has been the image techniques that enable the visualization of lipid distribution over a tissue with cell resolution. Thanks to the spatial information reported by such techniques, it is possible to associate a lipidome trait to individual cells, in fixed metabolic stages, which greatly facilitates understanding the metabolic changes associated to diverse pathological conditions, such as cancer. Furthermore, the image of lipids is becoming a kind of new molecular histology that has great chances to make an impact in the diagnostic units of the hospitals. Here, we examine the current state of the technology and analyze what the next steps to bring it into the diagnosis units should be. To illustrate the potential and challenges of this technology, we present a case study on clear cell renal cell carcinoma, a good model for analyzing malignant tumors due to their significant cellular and molecular heterogeneity.
Collapse
Affiliation(s)
- Ibai Calvo
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Barrio Sarriena S/N, 48940 Leioa, Spain
| | - Olatz Fresnedo
- Lipids&Liver, Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B. Sarriena, s/n, Leioa 48940, Spain
| | - Lorena Mosteiro
- Department of Pathology, Cruces University Hospital, 48903 Barakaldo, Spain
| | - José I López
- Biobizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Gorka Larrinaga
- Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; Department of Nursing, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B. Sarriena, s/n, Leioa 48940, Spain; Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B. Sarriena, s/n, Leioa 48940, Spain.
| | - José A Fernández
- Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Barrio Sarriena S/N, 48940 Leioa, Spain.
| |
Collapse
|
7
|
Wang H, Nikain C, Fortounas KI, Amengual J, Tufanli O, La Forest M, Yu Y, Wang MC, Watts R, Lehner R, Qiu Y, Cai M, Kurland IJ, Goldberg IJ, Rajan S, Hussain MM, Brodsky JL, Fisher EA. FITM2 deficiency results in ER lipid accumulation, ER stress, and reduced apolipoprotein B lipidation and VLDL triglyceride secretion in vitro and in mouse liver. Mol Metab 2024; 90:102048. [PMID: 39426520 PMCID: PMC11574801 DOI: 10.1016/j.molmet.2024.102048] [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: 07/26/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024] Open
Abstract
OBJECTIVE Triglycerides (TGs) associate with apolipoprotein B100 (apoB100) to form very low density lipoproteins (VLDLs) in the liver. The repertoire of factors that facilitate this association is incompletely understood. FITM2, an integral endoplasmic reticulum (ER) protein, was originally discovered as a factor participating in cytosolic lipid droplet (LD) biogenesis in tissues that do not form VLDL. We hypothesized that in the liver, in addition to promoting cytosolic LD formation, FITM2 would also transfer TG from its site of synthesis in the ER membrane to nascent VLDL particles within the ER lumen. METHODS Experiments were conducted using a rat hepatic cell line (McArdle-RH7777, or McA cells), an established model of mammalian lipoprotein metabolism, and mice. FITM2 expression was reduced using siRNA in cells and by liver specific cre-recombinase mediated deletion of the Fitm2 gene in mice. Effects of FITM2 deficiency on VLDL assembly and secretion in vitro and in vivo were measured by multiple methods, including density gradient ultracentrifugation, chromatography, mass spectrometry, stimulated Raman scattering (SRS) microscopy, sub-cellular fractionation, immunoprecipitation, immunofluorescence, and electron microscopy. MAIN FINDINGS 1) FITM2-deficient hepatic cells in vitro and in vivo secrete TG-depleted VLDL particles, but the number of particles is unchanged compared to controls; 2) FITM2 deficiency in mice on a high fat diet (HFD) results in decreased plasma TG levels. The number of apoB100-containing lipoproteins remains similar, but shift from VLDL to low density lipoprotein (LDL) density; 3) Both in vitro and in vivo, when TG synthesis is stimulated and FITM2 is deficient, TG accumulates in the ER, and despite its availability this pool is unable to fully lipidate apoB100 particles; 4) FITM2 deficiency disrupts ER morphology and results in ER stress. CONCLUSION The results suggest that FITM2 contributes to VLDL lipidation, especially when newly synthesized hepatic TG is in abundance. In addition to its fundamental importance in VLDL assembly, the results also suggest that under dysmetabolic conditions, FITM2 may be an important factor in the partitioning of TG between cytosolic LDs and VLDL particles.
Collapse
Affiliation(s)
- Haizhen Wang
- Department of Medicine (Cardiology), the Cardiovascular Research Center, and the Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NY, USA; College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Cyrus Nikain
- Department of Medicine (Cardiology), the Cardiovascular Research Center, and the Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NY, USA; Chemical Biology Program, Memorial Sloan Kettering Cancer Center and Weill Graduate School of Medical Sciences, Cornell University, NY, USA
| | - Konstantinos I Fortounas
- Department of Medicine (Cardiology), the Cardiovascular Research Center, and the Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NY, USA
| | - Jaume Amengual
- Department of Medicine (Cardiology), the Cardiovascular Research Center, and the Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NY, USA; Department of Food Sciences and Human Nutrition, University of Illinois, Urbana-Champaign, IL, USA
| | - Ozlem Tufanli
- Department of Medicine (Cardiology), the Cardiovascular Research Center, and the Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NY, USA
| | - Maxwell La Forest
- Department of Medicine (Cardiology), the Cardiovascular Research Center, and the Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NY, USA
| | - Yong Yu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA; State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Meng C Wang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA; Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Russell Watts
- Department of Pediatrics and Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - Richard Lehner
- Department of Pediatrics and Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - Yunping Qiu
- Stable Isotope and Metabolomics Core Facility, Albert Einstein College of Medicine, NY, USA
| | - Min Cai
- Stable Isotope and Metabolomics Core Facility, Albert Einstein College of Medicine, NY, USA
| | - Irwin J Kurland
- Stable Isotope and Metabolomics Core Facility, Albert Einstein College of Medicine, NY, USA
| | - Ira J Goldberg
- Department of Medicine (Endocrinology), NYU Grossman School of Medicine, NY, USA
| | - Sujith Rajan
- Department of Foundations of Medicine and Diabetes and Obesity Research Center, NYU Grossman Long Island School of Medicine, Mineola, NY, USA
| | - M Mahmood Hussain
- Department of Foundations of Medicine and Diabetes and Obesity Research Center, NYU Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences and the Center for Protein Conformational Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Edward A Fisher
- Department of Medicine (Cardiology), the Cardiovascular Research Center, and the Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NY, USA.
| |
Collapse
|
8
|
Kumar A, Yadav S, Choudhary V. The evolving landscape of ER-LD contact sites. Front Cell Dev Biol 2024; 12:1483902. [PMID: 39421023 PMCID: PMC11484260 DOI: 10.3389/fcell.2024.1483902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Lipid droplets (LDs) are evolutionarily conserved dynamic organelles that play an important role in cellular physiology. Growing evidence suggests that LD biogenesis occurs at discrete endoplasmic reticulum (ER) subdomains demarcated by the lipodystrophy protein, Seipin, lack of which impairs adipogenesis. However, the mechanisms of how these domains are selected is not completely known. These ER sites undergo ordered assembly of proteins and lipids to initiate LD biogenesis and facilitate establishment of ER-LD contact sites, a prerequisite for proper growth and maturation of droplets. LDs retain both physical and functional association with the ER throughout their lifecycle to facilitate bi-directional communication, such as exchange of proteins and lipids between the two organelles at these ER-LD contact sites. In recent years several molecular tethers have been identified that bridge ER and LDs together including few proteins that are found exclusively at these ER-LD contact interface. Here, we discuss recent advances in understanding the role of factors that ensure functionality of ER-LD contact site machinery for LD homeostasis.
Collapse
Affiliation(s)
| | | | - Vineet Choudhary
- Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| |
Collapse
|
9
|
Brodsky JL, Iyer A, Fortounas KI, Fisher EA. The emerging role of fat-inducing transcript 2 in endoplasmic reticulum proteostasis and lipoprotein biogenesis. Curr Opin Lipidol 2024; 35:248-252. [PMID: 39172716 PMCID: PMC11387134 DOI: 10.1097/mol.0000000000000943] [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] [Indexed: 08/24/2024]
Abstract
PURPOSE OF REVIEW This review examines the evolving role of the fat-inducing transcript 2 (FIT2) protein in lipid droplet (LD) biology and its broader implications in cellular physiology and disease. With recent advancements in understanding FIT2 function across various model systems, this review provides a timely synthesis of its mechanisms and physiological significance. RECENT FINDINGS FIT2, an endoplasmic reticulum (ER)-resident protein, has been established as a critical regulator of LD formation in diverse organisms, from yeast to mammals. It facilitates LD biogenesis by sequestering diacylglycerol (DAG) and potentially influencing ER membrane dynamics. Beyond its role in lipid metabolism, FIT2 intersects with the ER-associated degradation (ERAD), is critical for protein homeostasis, and is linked to the unfolded protein response (UPR). Dysregulation of FIT2 has also been linked to metabolic disorders such as insulin resistance and lipodystrophy, highlighting its clinical relevance. SUMMARY Insights into FIT2 function underscore its pivotal role in LD formation and lipid homeostasis. Understanding its involvement in ER proteostasis and very low density lipoprotein biogenesis has broad implications for metabolic diseases and cancer. Therapeutic strategies targeting FIT2 may offer novel approaches to modulate lipid metabolism and mitigate associated pathologies. Further research is needed to elucidate the full spectrum of FIT2's interactions within cellular lipid and protein networks, potentially uncovering new therapeutic avenues for metabolic and ER stress-related disorders.
Collapse
Affiliation(s)
- Jeffrey L. Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Protein Conformational Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anuradha Iyer
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Konstantinos I. Fortounas
- Division of Cardiology and the Department of Medicine, NYU School of Medicine, New York, NY, USA
- Cardiovascular Research Center and the Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, New York, NY, USA
| | - Edward A. Fisher
- Division of Cardiology and the Department of Medicine, NYU School of Medicine, New York, NY, USA
- Cardiovascular Research Center and the Marc and Ruti Bell Program in Vascular Biology, NYU School of Medicine, New York, New York, NY, USA
| |
Collapse
|
10
|
Klemm RW, Carvalho P. Lipid Droplets Big and Small: Basic Mechanisms That Make Them All. Annu Rev Cell Dev Biol 2024; 40:143-168. [PMID: 39356808 DOI: 10.1146/annurev-cellbio-012624-031419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Lipid droplets (LDs) are dynamic storage organelles with central roles in lipid and energy metabolism. They consist of a core of neutral lipids, such as triacylglycerol, which is surrounded by a monolayer of phospholipids and specialized surface proteins. The surface composition determines many of the LD properties, such as size, subcellular distribution, and interaction with partner organelles. Considering the diverse energetic and metabolic demands of various cell types, it is not surprising that LDs are highly heterogeneous within and between cell types. Despite their diversity, all LDs share a common biogenesis mechanism. However, adipocytes have evolved specific adaptations of these basic mechanisms, enabling the regulation of lipid and energy metabolism at both the cellular and organismal levels. Here, we discuss recent advances in the understanding of both the general mechanisms of LD biogenesis and the adipocyte-specific adaptations controlling these fascinating organelles.
Collapse
Affiliation(s)
- Robin W Klemm
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom;
| | - Pedro Carvalho
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom;
| |
Collapse
|
11
|
Graff J, Schneiter R. FIT2 proteins and lipid droplet emergence, an interplay between phospholipid synthesis, surface tension, and membrane curvature. Front Cell Dev Biol 2024; 12:1422032. [PMID: 38872930 PMCID: PMC11169642 DOI: 10.3389/fcell.2024.1422032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 05/06/2024] [Indexed: 06/15/2024] Open
Abstract
Lipid droplets (LDs) serve as intracellular compartments primarily dedicated to the storage of metabolic energy in the form of neutral lipids. The processes that regulate and control LD biogenesis are being studied extensively and are gaining significance due to their implications in major metabolic disorders, including type 2 diabetes and obesity. A protein of particular interest is Fat storage-Inducing Transmembrane 2 (FIT2), which affects the emergence step of LD biogenesis. Instead of properly emerging towards the cytosol, LDs in FIT2-deficient cells remain embedded within the membrane of the endoplasmic reticulum (ER). In vitro studies revealed the ability of FIT2 to bind both di- and triacylglycerol (DAG/TAG), key players in lipid storage, and its activity to cleave acyl-CoA. However, the translation of these in vitro functions to the observed embedding of LDs in FIT2 deficient cells remains to be established. To understand the role of FIT2 in vivo, we discuss the parameters that affect LD emergence. Our focus centers on the role that membrane curvature and surface tension play in LD emergence, as well as the impact that the lipid composition exerts on these key parameters. In addition, we discuss hypotheses on how FIT2 could function locally to modulate lipids at sites of LD emergence.
Collapse
Affiliation(s)
| | - Roger Schneiter
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
12
|
Deng Y, Zhu H, Wang Y, Dong Y, Du J, Yu Q, Li M. The Endoplasmic Reticulum-Plasma Membrane Tethering Protein Ice2 Controls Lipid Droplet Size via the Regulation of Phosphatidylcholine in Candida albicans. J Fungi (Basel) 2024; 10:87. [PMID: 38276033 PMCID: PMC10817647 DOI: 10.3390/jof10010087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/13/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Lipid droplets (LDs) are intracellular organelles that play important roles in cellular lipid metabolism; they change their sizes and numbers in response to both intracellular and extracellular signals. Changes in LD size reflect lipid synthesis and degradation and affect many cellular activities, including energy supply and membrane synthesis. Here, we focused on the function of the endoplasmic reticulum-plasma membrane tethering protein Ice2 in LD dynamics in the fungal pathogen Candida albicans (C. albicans). Nile red staining and size quantification showed that the LD size increased in the ice2Δ/Δ mutant, indicating the critical role of Ice2 in the regulation of LD dynamics. A lipid content analysis further demonstrated that the mutant had lower phosphatidylcholine levels. As revealed with GFP labeling and fluorescence microscopy, the methyltransferase Cho2, which is involved in phosphatidylcholine synthesis, had poorer localization in the plasma membrane in the mutant than in the wild-type strain. Interestingly, the addition of the phosphatidylcholine precursor choline led to the recovery of normal-sized LDs in the mutant. These results indicated that Ice2 regulates LD size by controlling intracellular phosphatidylcholine levels and that endoplasmic reticulum-plasma membrane tethering proteins play a role in lipid metabolism regulation in C. albicans. This study provides significant findings for further investigation of the lipid metabolism in fungi.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mingchun Li
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China; (Y.D.); (H.Z.); (Y.W.); (Y.D.); (J.D.)
| |
Collapse
|
13
|
Rudenskaya GE, Bostanova FM, Medvedeva AS, Lotnik EE, Chausova PA, Shchagina OA. [A case of rare hereditary Siddiqi syndrome with novel neuropsychiatric signs]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:171-176. [PMID: 39731388 DOI: 10.17116/jnevro2024124121171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2024]
Abstract
A fifth world case of autosomal recessive Siddiqi syndrome (SIDDIS) related to FITM2 gene is presented. In a consanguineous Lezgin (a Dagestan ethnicity) family, there were two affected brothers aged 28 yrs (proband, personally examined) and 32 yrs. Whole-exome sequencing followed by familial Sanger sequencing detected a novel FITM2 missence variant c.452A>G, p.Asp151Gly homozygous in both patients and heterozygous in parents and unaffected brother. SIDDIS typical features in the patients were physical and motor delay with neuropsychiatric regress, loss of speech and walking, early-onset progressive sensorineural deafness, cognitive deficiency, skin lesion. However, there was no dystonia, which is a characteristic SIDDIS feature, while at least the proband had spastic paraparesis not described in SIDDIS earlier. Another distinction was mental state: behavioral disorders in both patients, positive symptoms in the proband and severe cognitive impairment in the brother. The case expands our understanding of the clinical and molecular-genetic spectrum of SIDDIS.
Collapse
Affiliation(s)
- G E Rudenskaya
- Bochkov Research Centre for Medical Genetics, Moscow, Russia
| | - F M Bostanova
- Bochkov Research Centre for Medical Genetics, Moscow, Russia
| | | | - E E Lotnik
- Bochkov Research Centre for Medical Genetics, Moscow, Russia
| | - P A Chausova
- Bochkov Research Centre for Medical Genetics, Moscow, Russia
| | - O A Shchagina
- Bochkov Research Centre for Medical Genetics, Moscow, Russia
| |
Collapse
|
14
|
Cala SE, Carruthers NJ, Stemmer PM, Chen Z, Chen X. Activation of Ca 2+ transport in cardiac microsomes enriches functional sets of ER and SR proteins. Mol Cell Biochem 2024; 479:85-98. [PMID: 37036634 PMCID: PMC10786961 DOI: 10.1007/s11010-023-04708-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/12/2023] [Indexed: 04/11/2023]
Abstract
The importance of sarcoplasmic reticulum (SR) Ca2+-handling in heart has led to detailed understanding of Ca2+-release and re-uptake protein complexes, while less is known about other endoplasmic reticulum (ER) functions in the heart. To more fully understand cardiac SR and ER functions, we analyzed cardiac microsomes based on their increased density through the actions of the SR Ca2+-ATPase (SERCA) and the ryanodine receptor that are highly active in cardiomyocytes. Crude cardiac microsomal vesicles loaded with Ca oxalate produced two higher density subfractions, MedSR and HighSR. Proteins from 20.0 μg of MV, MedSR, and HighSR protein were fractionated using SDS-PAGE, then trypsinized from 20 separate gel pieces, and analyzed by LC-MS/MS to determine protein content. From 62,000 individual peptide spectra obtained, we identified 1105 different proteins, of which 354 were enriched ≥ 2.0-fold in SR fractions compared to the crude membrane preparation. Previously studied SR proteins were all enriched, as were proteins associated with canonical ER functions. Contractile, mitochondrial, and sarcolemmal proteins were not enriched. Comparing the levels of SERCA-positive SR proteins in MedSR versus HighSR vesicles produced a range of SR subfraction enrichments signifying differing levels of Ca2+ leak co-localized in the same membrane patch. All known junctional SR proteins were more enriched in MedSR, while canonical ER proteins were more enriched in HighSR membrane. Proteins constituting other putative ER/SR subdomains also exhibited average Esub enrichment values (mean ± S.D.) that spanned the range of possible Esub values, suggesting that functional sets of proteins are localized to the same areas of the ER/SR membrane. We conclude that active Ca2+ loading of cardiac microsomes, reflecting the combined activities of Ca2+ uptake by SERCA, and Ca2+ leak by RyR, permits evaluation of multiple functional ER/SR subdomains. Sets of proteins from these subdomains exhibited similar enrichment patterns across membrane subfractions, reflecting the relative levels of SERCA and RyR present within individual patches of cardiac ER and SR.
Collapse
Affiliation(s)
- Steven E Cala
- Department of Physiology, Wayne State University, Detroit, MI, 48201, USA.
| | | | - Paul M Stemmer
- Institute of Environmental Health Sciences, Wayne State University, Detroit, USA
| | - Zhenhui Chen
- Krannert Institute of Cardiology, Indiana University, Indianapolis, IN, USA
| | - Xuequn Chen
- Department of Physiology, Wayne State University, Detroit, MI, 48201, USA
| |
Collapse
|
15
|
Mondal S, Ghosh S. Liposome-Mediated Anti-Viral Drug Delivery Across Blood-Brain Barrier: Can Lipid Droplet Target Be Game Changers? Cell Mol Neurobiol 2023; 44:9. [PMID: 38123863 PMCID: PMC11407177 DOI: 10.1007/s10571-023-01443-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/02/2023] [Indexed: 12/23/2023]
Abstract
Lipid droplets (LDs) are subcellular organelles secreted from the endoplasmic reticulum (ER) that play a major role in lipid homeostasis. Recent research elucidates additional roles of LDs in cellular bioenergetics and innate immunity. LDs activate signaling cascades for interferon response and secretion of pro-inflammatory cytokines. Since balanced lipid homeostasis is critical for neuronal health, LDs play a crucial role in neurodegenerative diseases. RNA viruses enhance the secretion of LDs to support various phases of their life cycle in neurons which further leads to neurodegeneration. Targeting the excess LD formation in the brain could give us a new arsenal of antiviral therapeutics against neuroviruses. Liposomes are a suitable drug delivery system that could be used for drug delivery in the brain by crossing the Blood-Brain Barrier. Utilizing this, various pharmacological inhibitors and non-coding RNAs can be delivered that could inhibit the biogenesis of LDs or reduce their sizes, reversing the excess lipid-related imbalance in neurons. Liposome-Mediated Antiviral Drug Delivery Across Blood-Brain Barrier. Developing effective antiviral drug is challenging and it doubles against neuroviruses that needs delivery across the Blood-Brain Barrier (BBB). Lipid Droplets (LDs) are interesting targets for developing antivirals, hence targeting LD formation by drugs delivered using Liposomes can be game changers.
Collapse
Affiliation(s)
- Sourav Mondal
- CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Sourish Ghosh
- CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India.
| |
Collapse
|
16
|
Wang H, Nikain C, Amengual J, La Forest M, Yu Y, Wang MC, Watts R, Lehner R, Qiu Y, Cai M, Kurland IJ, Goldberg IJ, Rajan S, Hussain MM, Brodsky JL, Fisher EA. FITM2 deficiency results in ER lipid accumulation, ER stress, reduced apolipoprotein B lipidation, and VLDL triglyceride secretion in vitro and in mouse liver. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.05.570183. [PMID: 38106013 PMCID: PMC10723279 DOI: 10.1101/2023.12.05.570183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Objectives Triglyceride (TG) association with apolipoprotein B100 (apoB100) serves to form very low density lipoproteins (VLDL) in the liver. The repertoire of factors that facilitate this association is incompletely defined. FITM2, an integral endoplasmic reticulum (ER) protein, was originally discovered as a factor participating in cytoplasmic lipid droplets (LDs) in tissues that do not form VLDL. We hypothesized that in the liver, in addition to promoting cytosolic LD formation, FITM2 would also transfer TG from its site of synthesis in the ER membrane to nascent VLDL particles within the ER lumen. Methods Experiments were conducted using a rat hepatic cell line (McArdle-RH7777, or McA cells), an established model of mammalian lipoprotein metabolism, and mice. FITM2 expression was reduced using siRNA in cells and by liver specific cre-recombinase mediated deletion of the Fitm2 gene in mice. Effects of FITM2 deficiency on VLDL assembly and secretion in vitro and in vivo were measured by multiple methods, including density gradient ultracentrifugation, chromatography, mass spectrometry, simulated Raman spectroscopy (SRS) microscopy, sub-cellular fractionation, immunoprecipitation, immunofluorescence, and electron microscopy. Main findings 1) FITM2-deficient hepatic cells in vitro and in vivo secrete TG-depleted VLDL particles, but the number of particles is unchanged compared to controls; 2) FITM2 deficiency in mice on a high fat diet (HFD) results in decreased plasma TG levels. The number of apoB100-containing lipoproteins remains similar, but shift from VLDL to LDL density; 3) Both in vitro and in vivo , when TG synthesis is stimulated and FITM2 is deficient, TG accumulates in the ER, and despite its availability this pool is unable to fully lipidate apoB100 particles; 4) FITM2 deficiency disrupts ER morphology and results in ER stress. Principal conclusions The results suggest that FITM2 contributes to VLDL lipidation, especially when newly synthesized hepatic TG is in abundance. In addition to its fundamental importance in VLDL assembly, the results also suggest that under dysmetabolic conditions, FITM2 may be a limiting factor that ultimately contributes to non-alcoholic fatty liver disease (NAFLD) and steatohepatitis (NASH).
Collapse
|
17
|
Kumari RM, Khatri A, Chaudhary R, Choudhary V. Concept of lipid droplet biogenesis. Eur J Cell Biol 2023; 102:151362. [PMID: 37742390 PMCID: PMC7615795 DOI: 10.1016/j.ejcb.2023.151362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/26/2023] Open
Abstract
Lipid droplets (LD) are functionally conserved fat storage organelles found in all cell types. LDs have a unique structure comprising of a hydrophobic core of neutral lipids (fat), triacylglycerol (TAG) and cholesterol esters (CE) surrounded by a phospholipid monolayer. LD surface is decorated by a multitude of proteins and enzymes rendering this compartment functional. Accumulating evidence suggests that LDs originate from discrete ER-subdomains, demarcated by the lipodystrophy protein seipin, however, the mechanisms of which are not well understood. LD biogenesis factors together with biophysical properties of the ER membrane orchestrate spatiotemporal regulation of LD nucleation and growth at specific ER subdomains in response to metabolic cues. Defects in LD formation manifests in several human pathologies, including obesity, lipodystrophy, ectopic fat accumulation, and insulin resistance. Here, we review recent advances in understanding the molecular events during initial stages of eukaryotic LD assembly and discuss the critical role of factors that ensure fidelity of this process.
Collapse
Affiliation(s)
- R Mankamna Kumari
- Lipid Metabolism Laboratory, Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Amit Khatri
- Lipid Metabolism Laboratory, Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Ritika Chaudhary
- Lipid Metabolism Laboratory, Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Vineet Choudhary
- Lipid Metabolism Laboratory, Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India.
| |
Collapse
|
18
|
Moulistanos A, Nikolaou T, Sismanoglou S, Gkagkavouzis K, Karaiskou N, Antonopoulou E, Triantafyllidis A, Papakostas S. Investigating the role of genetic variation in vgll3 and six6 in the domestication of gilthead seabream ( Sparus aurata Linnaeus) and European seabass ( Dicentrarchus labrax Linnaeus). Ecol Evol 2023; 13:e10727. [PMID: 38020694 PMCID: PMC10654472 DOI: 10.1002/ece3.10727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Gene function conservation is crucial in molecular ecology, especially for key traits like growth and maturation in teleost fish. The vgll3 and six6 genes are known to influence age-at-maturity in Atlantic salmon, but their impact on other fish species is poorly understood. Here, we investigated the association of vgll3 and six6 in the domestication of gilthead seabream and European seabass, both undergoing selective breeding for growth-related traits in the Mediterranean. We analysed two different sets of samples using two different genotyping approaches. The first dataset comprised farmed and wild populations from Greece, genotyped for SNPs within the two genes ('gene-level genotyping'). The second dataset examined 300-600 k SNPs located in the chromosomes of the two genes, derived from a meta-analysis of a Pool-Seq experiment involving farmed and wild populations distributed widely across the Mediterranean ('chromosome-level genotyping'). The gene-level analysis revealed a statistically significant allele frequency differences between farmed and wild populations on both genes in each species. This finding was partially supported by the chromosome-level analysis, identifying highly differentiated regions may be involved in the domestication process at varying distances from the candidate genes. Noteworthy genomic features were found, such as a CpG island in gilthead seabream and novel candidate genes in European seabass, warranting further investigation. These findings support a putative role of vgll3 and six6 in the maturation and growth of gilthead seabream and European seabass, emphasizing the need for further research on their conserved function.
Collapse
Affiliation(s)
- Aristotelis Moulistanos
- Department of Genetics, Development & Molecular Biology, School of Biology, Faculty of ScienceAristotle University of ThessalonikiThessalonikiGreece
- Genomics and Epigenomics Translational Research (GENeTres)Center for Interdisciplinary Research and Innovation (CIRI‐AUTH), Balkan CenterThessalonikiGreece
| | - Theopisti Nikolaou
- Department of Genetics, Development & Molecular Biology, School of Biology, Faculty of ScienceAristotle University of ThessalonikiThessalonikiGreece
| | - Smaragda Sismanoglou
- Department of Genetics, Development & Molecular Biology, School of Biology, Faculty of ScienceAristotle University of ThessalonikiThessalonikiGreece
| | - Konstantinos Gkagkavouzis
- Department of Genetics, Development & Molecular Biology, School of Biology, Faculty of ScienceAristotle University of ThessalonikiThessalonikiGreece
- Genomics and Epigenomics Translational Research (GENeTres)Center for Interdisciplinary Research and Innovation (CIRI‐AUTH), Balkan CenterThessalonikiGreece
| | - Nikoleta Karaiskou
- Department of Genetics, Development & Molecular Biology, School of Biology, Faculty of ScienceAristotle University of ThessalonikiThessalonikiGreece
- Genomics and Epigenomics Translational Research (GENeTres)Center for Interdisciplinary Research and Innovation (CIRI‐AUTH), Balkan CenterThessalonikiGreece
| | - Efthimia Antonopoulou
- Department of Zoology, School of BiologyAristotle University of ThessalonikiThessalonikiGreece
| | - Alexandros Triantafyllidis
- Department of Genetics, Development & Molecular Biology, School of Biology, Faculty of ScienceAristotle University of ThessalonikiThessalonikiGreece
- Genomics and Epigenomics Translational Research (GENeTres)Center for Interdisciplinary Research and Innovation (CIRI‐AUTH), Balkan CenterThessalonikiGreece
| | - Spiros Papakostas
- Department of Science and TechnologyInternational Hellenic UniversityThessalonikiGreece
| |
Collapse
|
19
|
Naser I, Yabu Y, Maeda Y, Tanaka T. Highly Efficient Genetic Transformation Methods for the Marine Oleaginous Diatom Fistulifera solaris. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2023; 25:657-665. [PMID: 36512290 DOI: 10.1007/s10126-022-10189-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/27/2022] [Indexed: 06/17/2023]
Abstract
The oleaginous diatom Fistulifera solaris is a promising producer of biofuel owing to the high content of the lipids. A genetic transformation technique by microparticle bombardment for this diatom was already established. However, the transformation efficiency was significantly lower than those of other diatoms. Devoting efforts to advance the genetic modifications of this diatom is crucial to unlock its full potential. In this study, we optimized the microparticle bombardment protocol, and newly established a multi-pulse electroporation protocol for this diatom. The nutrient-rich medium in the pre-culture stage played an essential role to increase the transformation efficiency of the bombardment method. On the other hand, use of the nutrient-rich medium in the electroporation experiments resulted in decreasing the efficiency because excess nutrient salts could hamper to establish the best conductivity condition. Adjustments on the number and voltage of the poring pulses were also critical to obtain the best balance between cell viability and efficient pore formation. Under the optimized conditions, the transformation efficiencies of microparticle bombardment and multi-pulse electroporation were 111 and 82 per 108 cells, respectively (37 and 27 times higher than the conventional bombardment method). With the aid of the optimized protocol, we successfully developed the transformant clone over-expressing the endogenous fat storage-inducing transmembrane protein (FIT)-like protein, which was previously found in the genome of the oleaginous diatom F. solaris and the oleaginous eustigmatophyte Nannochloropsis gaditana. This study provides powerful techniques to investigate and further enhance the metabolic functions of F. solaris by genetic engineering.
Collapse
Affiliation(s)
- Insaf Naser
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, 184-8588, Koganei, Tokyo, Japan
| | - Yusuke Yabu
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, 184-8588, Koganei, Tokyo, Japan
| | - Yoshiaki Maeda
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, 184-8588, Koganei, Tokyo, Japan
- Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8572, Japan
| | - Tsuyoshi Tanaka
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, 184-8588, Koganei, Tokyo, Japan.
| |
Collapse
|
20
|
Yang C, Yang L, Yang L, Li S, Ye L, Ye J, Chen C, Zeng Y, Zhu M, Lin X, Peng Q, Wang Y, Jin M. Plasma Proteomics Study Between the Frequent Exacerbation and Infrequent Exacerbation Phenotypes of Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2023; 18:1713-1728. [PMID: 37581107 PMCID: PMC10423573 DOI: 10.2147/copd.s408361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 07/09/2023] [Indexed: 08/16/2023] Open
Abstract
Background Frequent exacerbation (FE) and infrequent exacerbation (IE) are two phenotypes of chronic obstructive pulmonary disease (COPD), of which FE is associated with a higher incidence of exacerbation and a serious threat to human health. Because the pathogenesis mechanisms of FE are unclear, this study aims to identify FE-related proteins in the plasma via proteomics for use as predictive, diagnostic, and therapeutic biomarkers of COPD. Methods A cross-sectional study was conducted in which plasma protein profiles were analyzed in COPD patients at stable stage, and differentially expressed proteins (DEPs) were screened out between the FE and IE patients. FE-related DEPs were identified using data-independent acquisition-based proteomics and bioinformatics analyses. In addition, FE-related candidates were verified by enzyme-linked immunosorbent assay. Results In this study, 47 DEPs were screened out between the FE and IE groups, including 20 upregulated and 27 downregulated proteins. Key biological functions (eg, neutrophil degranulation, extracellular exosome, protein homodimerization activity) and signaling pathways (eg, arginine and proline metabolism) were enriched in association with the FE phenotype. Receiver operating characteristic (ROC) analysis of the 11 combined DEPs revealed an area under the curve of 0.985 (p <0.05) for discriminating FE from IE. Moreover, correlation and ROC curve analyses indicated that creatine kinase, M-type (CKM) and fat storage-inducing transmembrane protein 1 (FITM1) might be clinically significant in patients with the FE phenotype. In addition, plasma expression levels of CKM and FITM1 were validated to be significantly decreased in the FE group compared with the IE group (CKM: p <0.01; FITM1: p <0.05). Conclusion In this study, novel insights into COPD pathogenesis were provided by investigating and comparing plasma protein profiles between the FE and IE patients. CKM, FITM1, and a combinative biomarker panel may serve as useful tools for assisting in the precision diagnosis and effective treatment of the FE phenotype of COPD.
Collapse
Affiliation(s)
- Chengyu Yang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Huadong Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
| | - Li Yang
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, People’s Republic of China
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, People’s Republic of China
| | - Lei Yang
- Longhua Innovation Institute for Biotechnology, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Shuiming Li
- Longhua Innovation Institute for Biotechnology, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Ling Ye
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, People’s Republic of China
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Jinfeng Ye
- Longhua Innovation Institute for Biotechnology, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Chengshui Chen
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, People’s Republic of China
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Zhejiang, 324000, People’s Republic of China
| | - Yiming Zeng
- Department of Pulmonary and Critical Care Medicine, the Second Affiliated Hospital of Fujian Medical University, Respiratory Medicine Center of Fujian Province, Quanzhou, Fujian, 362000, People’s Republic of China
| | - Mengchan Zhu
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Xiaoping Lin
- Department of Pulmonary and Critical Care Medicine, the Second Affiliated Hospital of Fujian Medical University, Respiratory Medicine Center of Fujian Province, Quanzhou, Fujian, 362000, People’s Republic of China
| | - Qing Peng
- Department of Pulmonary and Critical Care Medicine, Minhang Hospital, Fudan University, Shanghai, 201199, People’s Republic of China
| | - Yun Wang
- Longhua Innovation Institute for Biotechnology, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong, 515041, People’s Republic of China
| | - Meiling Jin
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, People’s Republic of China
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai, 200032, People’s Republic of China
| |
Collapse
|
21
|
Bombarda-Rocha V, Silva D, Badr-Eddine A, Nogueira P, Gonçalves J, Fresco P. Challenges in Pharmacological Intervention in Perilipins (PLINs) to Modulate Lipid Droplet Dynamics in Obesity and Cancer. Cancers (Basel) 2023; 15:4013. [PMID: 37568828 PMCID: PMC10417315 DOI: 10.3390/cancers15154013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
Perilipins (PLINs) are the most abundant proteins in lipid droplets (LD). These LD-associated proteins are responsible for upgrading LD from inert lipid storage structures to fully functional organelles, fundamentally integrated in the lipid metabolism. There are five distinct perilipins (PLIN1-5), each with specific expression patterns and metabolic activation, but all capable of regulating the activity of lipases on LD. This plurality creates a complex orchestrated mechanism that is directly related to the healthy balance between lipogenesis and lipolysis. Given the essential role of PLINs in the modulation of the lipid metabolism, these proteins can become interesting targets for the treatment of lipid-associated diseases. Since reprogrammed lipid metabolism is a recognized cancer hallmark, and obesity is a known risk factor for cancer and other comorbidities, the modulation of PLINs could either improve existing treatments or create new opportunities for the treatment of these diseases. Even though PLINs have not been, so far, directly considered for pharmacological interventions, there are many established drugs that can modulate PLINs activity. Therefore, the aim of this study is to assess the involvement of PLINs in diseases related to lipid metabolism dysregulation and whether PLINs can be viewed as potential therapeutic targets for cancer and obesity.
Collapse
Affiliation(s)
- Victória Bombarda-Rocha
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Dany Silva
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Allal Badr-Eddine
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
| | - Patrícia Nogueira
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Jorge Gonçalves
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Paula Fresco
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
22
|
Herrera-Moro Huitron L, De Jesús-González LA, Martínez-Castillo M, Ulloa-Aguilar JM, Cabello-Gutierrez C, Helguera-Repetto C, Garcia-Cordero J, León Juárez M. Multifaceted Nature of Lipid Droplets in Viral Interactions and Pathogenesis. Microorganisms 2023; 11:1851. [PMID: 37513023 PMCID: PMC10386712 DOI: 10.3390/microorganisms11071851] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/15/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Once regarded as inert organelles with limited and ill-defined roles, lipid droplets (LDs) have emerged as dynamic entities with multifaceted functions within the cell. Recent research has illuminated their pivotal role as primary energy reservoirs in the form of lipids, capable of being metabolized to meet cellular energy demands. Their high dynamism is underscored by their ability to interact with numerous cellular organelles, notably the endoplasmic reticulum (the site of LD genesis) and mitochondria, which utilize small LDs for energy production. Beyond their contribution to cellular bioenergetics, LDs have been associated with viral infections. Evidence suggests that viruses can co-opt LDs to facilitate their infection cycle. Furthermore, recent discoveries highlight the role of LDs in modulating the host's immune response. Observations of altered LD levels during viral infections suggest their involvement in disease pathophysiology, potentially through production of proinflammatory mediators using LD lipids as precursors. This review explores these intriguing aspects of LDs, shedding light on their multifaceted nature and implications in viral interactions and disease development.
Collapse
Affiliation(s)
- Luis Herrera-Moro Huitron
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico
| | | | - Macario Martínez-Castillo
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - José Manuel Ulloa-Aguilar
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico
| | - Carlos Cabello-Gutierrez
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Departamento de Investigación en Virología y Micología, Calzada de Tlalpan 4502, Belisario Domínguez, Tlalpan 14080, Mexico
| | - Cecilia Helguera-Repetto
- Laboratorio de Microbiología y Diagnóstico Molecular, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico
| | - Julio Garcia-Cordero
- Departamento de Biomedicina Molecular, Cinvestav, Av. IPN# 2508, Mexico City 07360, Mexico
| | - Moisés León Juárez
- Laboratorio de Virología Perinatal y Diseño Molecular de Antígenos y Biomarcadores, Departamento de Inmunobioquímica, Instituto Nacional de Perinatología, Mexico City 11000, Mexico
| |
Collapse
|
23
|
Dalen KT, Li Y. Regulation of lipid droplets and cholesterol metabolism in adrenal cortical cells. VITAMINS AND HORMONES 2023; 124:79-136. [PMID: 38408810 DOI: 10.1016/bs.vh.2023.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
The adrenal gland is composed of two distinctly different endocrine moieties. The interior medulla consists of neuroendocrine chromaffin cells that secrete catecholamines like adrenaline and noradrenaline, while the exterior cortex consists of steroidogenic cortical cells that produce steroid hormones, such as mineralocorticoids (aldosterone), glucocorticoids (cortisone and cortisol) and androgens. Synthesis of steroid hormones in cortical cells requires substantial amounts of cholesterol, which is the common precursor for steroidogenesis. Cortical cells may acquire cholesterol from de novo synthesis and uptake from circulating low- and high-density lipoprotein particles (LDL and HDL). As cholesterol is part of the plasma membrane in all mammalian cells and an important regulator of membrane fluidity, cellular levels of free cholesterol are tightly regulated. To ensure a robust supply of cholesterol for steroidogenesis and to avoid cholesterol toxicity, cortical cells store large amounts of cholesterol as cholesteryl esters in intracellular lipid droplets. Cortical steroidogenesis relies on both mobilization of cholesterol from lipid droplets and constant uptake of circulating cholesterol to replenish lipid droplet stores. This chapter will describe mechanisms involved in cholesterol uptake, cholesteryl ester synthesis, lipid droplet formation, hydrolysis of stored cholesteryl esters, as well as their impact on steroidogenesis. Additionally, animal models and human diseases characterized by altered cortical cholesteryl ester storage, with or without abnormal steroidogenesis, will be discussed.
Collapse
Affiliation(s)
- Knut Tomas Dalen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Norway; The Norwegian Transgenic Center, Institute of Basic Medical Sciences, University of Oslo, Norway.
| | - Yuchuan Li
- Department of Hepato-Pancreato-Biliary Surgery, Institute of Clinical Medicine, University of Oslo, Norway
| |
Collapse
|
24
|
Bond LM, Ibrahim A, Lai ZW, Walzem RL, Bronson RT, Ilkayeva OR, Walther TC, Farese RV. Fitm2 is required for ER homeostasis and normal function of murine liver. J Biol Chem 2023; 299:103022. [PMID: 36805337 PMCID: PMC10027564 DOI: 10.1016/j.jbc.2023.103022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/17/2023] Open
Abstract
The endoplasmic reticulum (ER)-resident protein fat storage-inducing transmembrane protein 2 (FIT2) catalyzes acyl-CoA cleavage in vitro and is required for ER homeostasis and normal lipid storage in cells. The gene encoding FIT2 is essential for the viability of mice and worms. Whether FIT2 acts as an acyl-CoA diphosphatase in vivo and how this activity affects the liver, where the protein was discovered, are unknown. Here, we report that hepatocyte-specific Fitm2 knockout (FIT2-LKO) mice fed a chow diet exhibited elevated acyl-CoA levels, ER stress, and signs of liver injury. These mice also had more triglycerides in their livers than control littermates due, in part, to impaired secretion of triglyceride-rich lipoproteins and reduced capacity for fatty acid oxidation. We found that challenging FIT2-LKO mice with a high-fat diet worsened hepatic ER stress and liver injury but unexpectedly reversed the steatosis phenotype, similar to what is observed in FIT2-deficient cells loaded with fatty acids. Our findings support the model that FIT2 acts as an acyl-CoA diphosphatase in vivo and is crucial for normal hepatocyte function and ER homeostasis in the murine liver.
Collapse
Affiliation(s)
- Laura M Bond
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA; Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Ayon Ibrahim
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA; Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Zon W Lai
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA; Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA; Harvard T.H. Chan Advanced Multi-omics Platform, Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Rosemary L Walzem
- Department of Poultry Science and Graduate Faculty of Nutrition, Kleberg Animal & Food Science Center, Texas A&M University, College Station, Texas, USA
| | - Roderick T Bronson
- Rodent Histopathology Core, Harvard Medical School, Boston, Massachusetts, USA
| | - Olga R Ilkayeva
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke Molecular Physiology Institute, Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Tobias C Walther
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA; Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA; Harvard T.H. Chan Advanced Multi-omics Platform, Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA; Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA; Howard Hughes Medical Institute, Boston, Massachusetts, USA.
| | - Robert V Farese
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA; Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA; Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA.
| |
Collapse
|
25
|
Effects of the CDC10 ( Septin 7) Gene on the Proliferation and Differentiation of Bovine Intramuscular Preadipocyte and 3T3-L1 Cells. Animals (Basel) 2023; 13:ani13040609. [PMID: 36830396 PMCID: PMC9951720 DOI: 10.3390/ani13040609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/01/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Intramuscular fat content and marbling affecting meat quality are important economic traits in beef cattle. CDC10 (cell division cycle 10 or Septin 7), a member of the septin family involved in cellular proliferation, was considered as a functional and positional candidate gene for beef marbling. In a previous study, we revealed that the expression levels of CDC10 were also positively correlated with marbling scores in Japanese Black cattle. However, the regulatory mechanism of the CDC10 gene on IMF deposition in cattle remains unclear. In the present study, flow cytometry, EdU proliferation assays, and Oil Red O staining results showed that overexpression of CDC10 could promote the differentiation of bovine intramuscular preadipocyte (BIMP) and 3T3-L1 cells, whereas knockdown of CDC10 resulted in the opposite consequences. Furthermore, quantitative PCR and Western blotting results showed that overexpression of CDC10 could promote the expression levels of adipogenic marker genes PPARγ and C/EBPα at both mRNA and protein levels in BIMP and 3T3-L1 cells, whereas knockdown of CDC10 resulted in the opposite consequences. Our results provide new insights into the regulatory roles of CDC10 in adipocytes in animals.
Collapse
|
26
|
Cala SE, Carruthers NJ, Stemmer PM, Chen Z, Chen X. Activation of Ca transport in cardiac microsomes enriches functional sets of ER and SR proteins. RESEARCH SQUARE 2023:rs.3.rs-2557992. [PMID: 36798315 PMCID: PMC9934757 DOI: 10.21203/rs.3.rs-2557992/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The importance of sarcoplasmic reticulum (SR) Ca-handling in heart has led to detailed understanding of Ca-release and re-uptake protein complexes, while less is known about other endoplasmic reticulum (ER) functions in the heart. To more fully understand cardiac SR and ER functions, we analyzed cardiac microsomes based on their increased density through the actions of the SR Ca-ATPase (SERCA) and the ryanodine receptor that are highly active in cardiomyocytes. Crude cardiac microsomal vesicles loaded with Ca oxalate produced two higher density subfractions, MedSR and HighSR. Analyses of protein enrichments from the 3 membrane preparations (crude microsomes, MedSR, and HighSR), showed that only a third of microsomal proteins in heart, or 354 proteins, were enriched ≥2.0-fold in SR. Previously studied SR proteins were all enriched, as were proteins associated with canonical ER functions. Contractile, mitochondrial, and sarcolemmal proteins were not enriched. Comparing the levels of SERCA-positive SR proteins in MedSR versus HighSR vesicles produced a range of SR subfraction enrichments signifying differing levels of Ca leak (ryanodine receptor) co-localized in the same membrane patch. All known junctional SR proteins were more enriched in MedSR, while canonical ER proteins were more enriched in HighSR membrane. Proteins from other putative ER/SR subdomains also showed characteristic distributions among SR subpopulations. We conclude that active Ca loading of cardiac microsomes, reflecting the combined activities of Ca uptake by SERCA, and Ca leak by RyR, permits evaluation of multiple functional ER/SR subdomains. Sets of proteins from these subdomains exhibited similar enrichment patterns across membrane subfractions, reflecting the relative levels of SERCA and RyR present within individual patches of cardiac ER and SR.
Collapse
|
27
|
Dressler EA, Shaffer W, Bruno K, Krehbiel CR, Calvo-Lorenzo M, Richards CJ, Place SE, DeSilva U, Kuehn LA, Weaber RL, Bormann JM, Rolf MM. Heritability and variance component estimation for feed and water intake behaviors of feedlot cattle. J Anim Sci 2023; 101:skad386. [PMID: 37967310 PMCID: PMC10699840 DOI: 10.1093/jas/skad386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 11/14/2023] [Indexed: 11/17/2023] Open
Abstract
Feed and water intake are two important aspects of cattle production that greatly impact the profitability, efficiency, and sustainability of producers. Feed and, to a lesser degree, water intake have been studied previously; however, there is little research on their associated animal behaviors and there is a lack of standardized phenotypes for these behaviors. Feed and water intakes obtained with an Insentec system (Hokofarm Group, The Netherlands) from 830 crossbred steers were used to compute five intake behaviors for both feed and water: daily sessions (DS), intake rate (IR), session size (SS), time per session (TS), and session interval (SI). Variance components and heritabilities were estimated for each trait. Heritabilities for feed intake behaviors were 0.50 ± 0.12, 0.63 ± 0.12, 0.40 ± 0.13, 0.35 ± 0.12, and 0.60 ± 0.12 for DS, IR, SS, TS, and SI, respectively. Heritabilities for water intake behaviors were 0.56 ± 0.11, 0.88 ± 0.07, 0.70 ± 0.11, 0.54 ± 0.12, and 0.80 ± 0.10 for NS, IR, SS, TS, and SI, respectively. Daily dry matter intake (DDMI) and daily water intake (DWI) had heritabilities of 0.57 ± 0.11 and 0.44 ± 0.11. Phenotypic correlations varied between pairs of traits (-0.83 to 0.82). Genetic correlations between DDMI and feed intake behaviors were moderate to high, while genetic correlations between DWI and water intake behaviors were low to moderate. Several significant single nucleotide polymorphisms (SNP) were identified for the feed and water intake behaviors. Genes and previously reported quantitative trait loci near significant SNPs were evaluated. The results indicated that feed and water intake behaviors are influenced by genetic factors and are heritable, providing one additional route to evaluate or manipulate feed and water intake.
Collapse
Affiliation(s)
- Elizabeth A Dressler
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS 66506, USA
| | - William Shaffer
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS 66506, USA
| | - Kelsey Bruno
- Department of Animal Science, Oklahoma State University, Stillwater, OK 74078, USA
| | - Clint R Krehbiel
- Department of Animal Science, Oklahoma State University, Stillwater, OK 74078, USA
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Michelle Calvo-Lorenzo
- Department of Animal Science, Oklahoma State University, Stillwater, OK 74078, USA
- Farm Animal Business, Elanco Animal Health, Greenfield, IN 46140, USA
| | - Chris J Richards
- Department of Animal Science, Oklahoma State University, Stillwater, OK 74078, USA
| | - Sara E Place
- Department of Animal Science, Oklahoma State University, Stillwater, OK 74078, USA
- Department of Animal Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Udaya DeSilva
- Department of Animal Science, Oklahoma State University, Stillwater, OK 74078, USA
| | - Larry A Kuehn
- USDA, ARS, Roman L. Hruska U.S. Meat Animal Research Center, Clay Center, NE 68933, USA
| | - Robert L Weaber
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS 66506, USA
| | - Jennifer M Bormann
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS 66506, USA
| | - Megan M Rolf
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
28
|
Bonar M, Anderson SJ, Anderson CR, Wittemyer G, Northrup JM, Shafer ABA. Genomic correlates for migratory direction in a free-ranging cervid. Proc Biol Sci 2022; 289:20221969. [PMID: 36475444 PMCID: PMC9727677 DOI: 10.1098/rspb.2022.1969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Animal migrations are some of the most ubiquitous and one of the most threatened ecological processes globally. A wide range of migratory behaviours occur in nature, and this behaviour is not uniform among and within species, where even individuals in the same population can exhibit differences. While the environment largely drives migratory behaviour, it is necessary to understand the genetic mechanisms influencing migration to elucidate the potential of migratory species to cope with novel conditions and adapt to environmental change. In this study, we identified genes associated with a migratory trait by undertaking pooled genome-wide scans on a natural population of migrating mule deer. We identified genomic regions associated with variation in migratory direction, including FITM1, a gene linked to the formation of lipids, and DPPA3, a gene linked to epigenetic modifications of the maternal line. Such a genetic basis for a migratory trait contributes to the adaptive potential of the species and might affect the flexibility of individuals to change their behaviour in the face of changes in their environment.
Collapse
Affiliation(s)
- Maegwin Bonar
- Environmental & Life Sciences Graduate Program, Trent University, Peterborough, Ontario, Canada K9L 0G2
| | - Spencer J. Anderson
- Environmental & Life Sciences Graduate Program, Trent University, Peterborough, Ontario, Canada K9L 0G2
| | - Charles R. Anderson
- Mammals Research Section, Colorado Parks and Wildlife, Fort Collins, CO 80523, USA
| | - George Wittemyer
- Department of Fish, Wildlife and Conservation Biology, Colorado State University, Fort Collins, CO 80523, USA
| | - Joseph M. Northrup
- Environmental & Life Sciences Graduate Program, Trent University, Peterborough, Ontario, Canada K9L 0G2,Wildlife Research and Monitoring Section, Ontario Ministry of Natural Resources & Forestry, Peterborough, Ontario, Canada K9J 3C7
| | - Aaron B. A. Shafer
- Environmental & Life Sciences Graduate Program, Trent University, Peterborough, Ontario, Canada K9L 0G2
| |
Collapse
|
29
|
The Theory of Carcino-Evo-Devo and Its Non-Trivial Predictions. Genes (Basel) 2022; 13:genes13122347. [PMID: 36553613 PMCID: PMC9777766 DOI: 10.3390/genes13122347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 12/04/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
To explain the sources of additional cell masses in the evolution of multicellular organisms, the theory of carcino-evo-devo, or evolution by tumor neofunctionalization, has been developed. The important demand for a new theory in experimental science is the capability to formulate non-trivial predictions which can be experimentally confirmed. Several non-trivial predictions were formulated using carcino-evo-devo theory, four of which are discussed in the present paper: (1) The number of cellular oncogenes should correspond to the number of cell types in the organism. The evolution of oncogenes, tumor suppressor and differentiation gene classes should proceed concurrently. (2) Evolutionarily new and evolving genes should be specifically expressed in tumors (TSEEN genes). (3) Human orthologs of fish TSEEN genes should acquire progressive functions connected with new cell types, tissues and organs. (4) Selection of tumors for new functions in the organism is possible. Evolutionarily novel organs should recapitulate tumor features in their development. As shown in this paper, these predictions have been confirmed by the laboratory of the author. Thus, we have shown that carcino-evo-devo theory has predictive power, fulfilling a fundamental requirement for a new theory.
Collapse
|
30
|
Wang G, Chen A, Wu Y, Wang D, Chang C, Yu G. Fat storage-inducing transmembrane proteins: beyond mediating lipid droplet formation. Cell Mol Biol Lett 2022; 27:98. [DOI: 10.1186/s11658-022-00391-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/23/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractFat storage-inducing transmembrane proteins (FITMs) were initially identified in 2007 as members of a conserved endoplasmic reticulum (ER) resident transmembrane protein gene family, and were found to be involved in lipid droplet (LD) formation. Recently, several studies have further demonstrated that the ability of FITMs to directly bind to triglyceride and diacylglycerol, and the diphosphatase activity of hydrolyzing fatty acyl-CoA, might enable FITMs to maintain the formation of lipid droplets, engage in lipid metabolism, and protect against cellular stress. Based on the distribution of FITMs in tissues and their important roles in lipid droplet biology and lipid metabolism, it was discovered that FITMs were closely related to muscle development, adipocyte differentiation, and energy metabolism. Accordingly, the abnormal expression of FITMs was not only associated with type 2 diabetes and lipodystrophy, but also with cardiac disease and several types of cancer. This study reviews the structure, distribution, expression regulation, and functionality of FITMs and their potential relationships with various metabolic diseases, hoping to provide inspiration for fruitful research directions and applications of FITM proteins. Moreover, this review will provide an important theoretical basis for the application of FITMs in the diagnosis and treatment of related diseases.
Collapse
|
31
|
Renne MF, Corey RA, Ferreira JV, Stansfeld PJ, Carvalho P. Seipin concentrates distinct neutral lipids via interactions with their acyl chain carboxyl esters. J Cell Biol 2022; 221:e202112068. [PMID: 35938957 PMCID: PMC9365673 DOI: 10.1083/jcb.202112068] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 05/28/2022] [Accepted: 07/12/2022] [Indexed: 11/22/2022] Open
Abstract
Lipid droplets (LDs) are essential for cellular lipid homeostasis by storing diverse neutral lipids (NLs), such as triacylglycerol (TAG), steryl esters (SE), and retinyl esters (RE). A proper assembly of TAG-containing LDs at the ER requires Seipin, a conserved protein often mutated in lipodystrophies. Here, we show that the yeast Seipin Sei1 and its partner Ldb16 also promote the storage of other NL in LDs. Importantly, this role of Sei1/Ldb16 is evolutionarily conserved as expression of human-Seipin restored normal SE-containing LDs in yeast Seipin mutants. As in the case of TAG, the formation of SE-containing LDs requires interactions between hydroxyl-residues in human Seipin or yeast Ldb16 with NL carboxyl esters. These findings provide a universal mechanism for Seipin-mediated LD formation and suggest a model for how Seipin distinguishes NLs from aliphatic phospholipid acyl chains in the center of the membrane bilayer.
Collapse
Affiliation(s)
- Mike F. Renne
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Robin A. Corey
- Department of Biochemistry, University of Oxford, Oxford, UK
| | | | - Phillip J. Stansfeld
- Department of Biochemistry, University of Oxford, Oxford, UK
- School of Life Sciences and Department of Chemistry, University of Warwick, Coventry, UK
| | - Pedro Carvalho
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| |
Collapse
|
32
|
Apte MS, Joshi AS. Membrane shaping proteins, lipids, and cytoskeleton: Recipe for nascent lipid droplet formation. Bioessays 2022; 44:e2200038. [PMID: 35832014 DOI: 10.1002/bies.202200038] [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: 02/15/2022] [Revised: 06/26/2022] [Accepted: 06/29/2022] [Indexed: 11/06/2022]
Abstract
Lipid droplets (LDs) are ubiquitous, neutral lipidorganelles that act as hubs of metabolic processes. LDs are structurally unique with a hydrophobic core that mainly consists of neutral lipids, sterol esters, and triglycerides, enclosed within a phospholipid monolayer. Nascent LD formation begins with the accumulation of neutral lipids in the endoplasmic reticulum (ER) bilayer. The ER membrane proteins such as seipin, LDAF1, FIT, and MCTPs are reported to play an important role in the formation of nascent LDs. As the LDs grow, they unmix from the highly charged ER membrane to form mature LDs. LD biogenesis is an exciting, emerging research area, and herein, we discuss the recent progress in our understanding of the formation of eukaryotic nascent LDs. We focus on the role of ER membrane shaping proteins such as reticulons and reticulon-like proteins, membrane lipids, and cytoskeleton proteins such as septin in the formation of nascent LDs.
Collapse
Affiliation(s)
- Manasi S Apte
- Department of Biochemistry & Cell and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| | - Amit S Joshi
- Department of Biochemistry & Cell and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
33
|
Ghanem M, Lewis GF, Xiao C. Recent advances in cytoplasmic lipid droplet metabolism in intestinal enterocyte. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159197. [PMID: 35820577 DOI: 10.1016/j.bbalip.2022.159197] [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: 12/24/2021] [Revised: 06/03/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022]
Abstract
Processing of dietary fats in the intestine is a highly regulated process that influences whole-body energy homeostasis and multiple physiological functions. Dysregulated lipid handling in the intestine leads to dyslipidemia and atherosclerotic cardiovascular disease. In intestinal enterocytes, lipids are incorporated into lipoproteins and cytoplasmic lipid droplets (CLDs). Lipoprotein synthesis and CLD metabolism are inter-connected pathways with multiple points of regulation. This review aims to highlight recent advances in the regulatory mechanisms of lipid processing in the enterocyte, with particular focus on CLDs. In-depth understanding of the regulation of lipid metabolism in the enterocyte may help identify therapeutic targets for the treatment and prevention of metabolic disorders.
Collapse
Affiliation(s)
- Murooj Ghanem
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Gary F Lewis
- Departments of Medicine and Physiology, University of Toronto, and University Health Network, Toronto, ON, Canada
| | - Changting Xiao
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
34
|
Lu H, Lei X, Winkler R, John S, Kumar D, Li W, Alnouti Y. Crosstalk of hepatocyte nuclear factor 4a and glucocorticoid receptor in the regulation of lipid metabolism in mice fed a high-fat-high-sugar diet. Lipids Health Dis 2022; 21:46. [PMID: 35614477 PMCID: PMC9134643 DOI: 10.1186/s12944-022-01654-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 05/06/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Hepatocyte nuclear factor 4α (HNF4α) and glucocorticoid receptor (GR), master regulators of liver metabolism, are down-regulated in fatty liver diseases. The present study aimed to elucidate the role of down-regulation of HNF4α and GR in fatty liver and hyperlipidemia. METHODS Adult mice with liver-specific heterozygote (HET) and knockout (KO) of HNF4α or GR were fed a high-fat-high-sugar diet (HFHS) for 15 days. Alterations in hepatic and circulating lipids were determined with analytical kits, and changes in hepatic mRNA and protein expression in these mice were quantified by real-time PCR and Western blotting. Serum and hepatic levels of bile acids were quantified by LC-MS/MS. The roles of HNF4α and GR in regulating hepatic gene expression were determined using luciferase reporter assays. RESULTS Compared to HFHS-fed wildtype mice, HNF4α HET mice had down-regulation of lipid catabolic genes, induction of lipogenic genes, and increased hepatic and blood levels of lipids, whereas HNF4α KO mice had fatty liver but mild hypolipidemia, down-regulation of lipid-efflux genes, and induction of genes for uptake, synthesis, and storage of lipids. Serum levels of chenodeoxycholic acid and deoxycholic acid tended to be decreased in the HNF4α HET mice but dramatically increased in the HNF4α KO mice, which was associated with marked down-regulation of cytochrome P450 7a1, the rate-limiting enzyme for bile acid synthesis. Hepatic mRNA and protein expression of sterol-regulatory-element-binding protein-1 (SREBP-1), a master lipogenic regulator, was induced in HFHS-fed HNF4α HET mice. In reporter assays, HNF4α cooperated with the corepressor small heterodimer partner to potently inhibit the transactivation of mouse and human SREBP-1C promoter by liver X receptor. Hepatic nuclear GR proteins tended to be decreased in the HNF4α KO mice. HFHS-fed mice with liver-specific KO of GR had increased hepatic lipids and induction of SREBP-1C and PPARγ, which was associated with a marked decrease in hepatic levels of HNF4α proteins in these mice. In reporter assays, GR and HNF4α synergistically/additively induced lipid catabolic genes. CONCLUSIONS induction of lipid catabolic genes and suppression of lipogenic genes by HNF4α and GR may mediate the early resistance to HFHS-induced fatty liver and hyperlipidemia.
Collapse
Affiliation(s)
- Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA.
| | - Xiaohong Lei
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Rebecca Winkler
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Savio John
- Department of Medicine, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Devendra Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Wenkuan Li
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yazen Alnouti
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
35
|
Schettini GP, Peripolli E, Alexandre PA, dos Santos WB, Pereira ASC, de Albuquerque LG, Baldi F, Curi RA. Transcriptome Profile Reveals Genetic and Metabolic Mechanisms Related to Essential Fatty Acid Content of Intramuscular Longissimus thoracis in Nellore Cattle. Metabolites 2022; 12:metabo12050471. [PMID: 35629975 PMCID: PMC9144777 DOI: 10.3390/metabo12050471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 05/05/2022] [Accepted: 05/16/2022] [Indexed: 02/05/2023] Open
Abstract
Beef is a source of essential fatty acids (EFA), linoleic (LA) and alpha-linolenic (ALA) acids, which protect against inflammatory and cardiovascular diseases in humans. However, the intramuscular EFA profile in cattle is a complex and polygenic trait. Thus, this study aimed to identify potential regulatory genes of the essential fatty acid profile in Longissimus thoracis of Nellore cattle finished in feedlot. Forty-four young bulls clustered in four groups of fifteen animals with extreme values for each FA were evaluated through differentially expressed genes (DEG) analysis and two co-expression methodologies (WGCNA and PCIT). We highlight the ECHS1, IVD, ASB5, and ERLIN1 genes and the TF NFIA, indicated in both FA. Moreover, we associate the NFYA, NFYB, PPARG, FASN, and FADS2 genes with LA, and the RORA and ELOVL5 genes with ALA. Furthermore, the functional enrichment analysis points out several terms related to FA metabolism. These findings contribute to our understanding of the genetic mechanisms underlying the beef EFA profile in Nellore cattle finished in feedlot.
Collapse
Affiliation(s)
- Gustavo Pimenta Schettini
- School of Agricultural and Veterinary Sciences, São Paulo State University, Jaboticabal 14884-900, SP, Brazil; (W.B.d.S.); (L.G.d.A.); (F.B.)
- Correspondence:
| | - Elisa Peripolli
- School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga 13635-900, SP, Brazil; (E.P.); (A.S.C.P.)
| | - Pâmela Almeida Alexandre
- Commonwealth Scientific and Industrial Research Organization, Agriculture & Food, St Lucia, QLD 4067, Australia;
| | - Wellington Bizarria dos Santos
- School of Agricultural and Veterinary Sciences, São Paulo State University, Jaboticabal 14884-900, SP, Brazil; (W.B.d.S.); (L.G.d.A.); (F.B.)
| | - Angélica Simone Cravo Pereira
- School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga 13635-900, SP, Brazil; (E.P.); (A.S.C.P.)
| | - Lúcia Galvão de Albuquerque
- School of Agricultural and Veterinary Sciences, São Paulo State University, Jaboticabal 14884-900, SP, Brazil; (W.B.d.S.); (L.G.d.A.); (F.B.)
| | - Fernando Baldi
- School of Agricultural and Veterinary Sciences, São Paulo State University, Jaboticabal 14884-900, SP, Brazil; (W.B.d.S.); (L.G.d.A.); (F.B.)
| | - Rogério Abdallah Curi
- School of Veterinary Medicine and Animal Science, São Paulo State University, Botucatu 18618-681, SP, Brazil;
| |
Collapse
|
36
|
Metabolic Engineering Strategies for Improved Lipid Production and Cellular Physiological Responses in Yeast Saccharomyces cerevisiae. J Fungi (Basel) 2022; 8:jof8050427. [PMID: 35628683 PMCID: PMC9144191 DOI: 10.3390/jof8050427] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/13/2022] [Accepted: 04/18/2022] [Indexed: 02/01/2023] Open
Abstract
Microbial lipids have been a hot topic in the field of metabolic engineering and synthetic biology due to their increased market and important applications in biofuels, oleochemicals, cosmetics, etc. This review first compares the popular hosts for lipid production and explains the four modules for lipid synthesis in yeast, including the fatty acid biosynthesis module, lipid accumulation module, lipid sequestration module, and fatty acid modification module. This is followed by a summary of metabolic engineering strategies that could be used for enhancing each module for lipid production. In addition, the efforts being invested in improving the production of value-added fatty acids in engineered yeast, such as cyclopropane fatty acid, ricinoleic acid, gamma linoleic acid, EPA, and DHA, are included. A discussion is further made on the potential relationships between lipid pathway engineering and consequential changes in cellular physiological properties, such as cell membrane integrity, intracellular reactive oxygen species level, and mitochondrial membrane potential. Finally, with the rapid development of synthetic biology tools, such as CRISPR genome editing tools and machine learning models, this review proposes some future trends that could be employed to engineer yeast with enhanced intracellular lipid production while not compromising much of its cellular health.
Collapse
|
37
|
Kozlov AP. Mammalian tumor-like organs. 2. Mammalian adipose has many tumor features and obesity is a tumor-like process. Infect Agent Cancer 2022; 17:15. [PMID: 35395810 PMCID: PMC8994355 DOI: 10.1186/s13027-022-00423-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 03/03/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND In previous publications, the author developed the theory of carcino-evo-devo, which predicts that evolutionarily novel organs should recapitulate some features of tumors in their development. MAIN TEXT Mammalian adipose is currently recognized as a multi-depot metabolic and endocrine organ consisting of several adipose tissues. Although lipid-storing cells and proteins are ancient, the adipose organ as a whole is evolutionarily novel to mammals. The adipose expansion has remarkable similarities with the growth of solid tumors. These similarities are the following: (1) The capability to unlimited expansion; (2) Reversible plasticity; (3) Induction of angiogenesis; (4) Chronic inflammation; (5) Remodeling and disfunction; (6) Systemic influence on the organism; (7) Hormone production; (8) Production of miRNAs that influence other tissues; (9) Immunosuppression; (10) DNA damage and resistance to apoptosis; (11) Destructive infiltration in other organs and tissues. These similarities include the majority of "hallmarks of cancer". In addition, lipomas are the most frequent soft tissue tumors, and similar drugs may be used for the treatment of obesity and cancer by preventing infiltration. This raises the possibility that obesity, at least in part, may represent an oncological problem. The existing similarities between adipose and tumors suggest the possible evolutionary origin of mammalian adipose from some ancestral benign mesenchymal hereditary tumors. Indeed, using a transgenic inducible zebrafish tumor model, we described many genes, which originated in fish and were expressed in fish tumors. Their human orthologs LEP, NOTCH1, SPRY1, PPARG, ID2, and CIDEA acquired functions connected with the adipose organ. They are also involved in tumor development in humans. CONCLUSION If the hypothesis of the evolutionary origin of the adipose organ from the ancestral hereditary tumor is correct, it may open new opportunities to resolve the oncological problem and the problem of the obesity epidemic. New interventions targeting LEP, NOTCH1, SPRY1, PPARG, ID2, and CIDEA gene network, in addition to what already is going on, can be designed for treatment and prevention of both obesity and tumors.
Collapse
Affiliation(s)
- A P Kozlov
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 3, Gubkina Street, Moscow, Russia, 117971.
- Peter the Great St. Petersburg Polytechnic University, 29, Polytekhnicheskaya Street, St. Petersburg, Russia, 195251.
- The Biomedical Center, 8, Viborgskaya Street, St. Petersburg, Russia, 194044.
| |
Collapse
|
38
|
Schettini GP, Peripolli E, Alexandre PA, Dos Santos WB, da Silva Neto JB, Pereira ASC, de Albuquerque LG, Curi RA, Baldi F. Transcriptomic profile of longissimus thoracis associated with fatty acid content in Nellore beef cattle. Anim Genet 2022; 53:264-280. [PMID: 35384007 DOI: 10.1111/age.13199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/25/2021] [Accepted: 03/24/2022] [Indexed: 11/29/2022]
Abstract
The beef fatty acid (FA) profile has the potential to impact human health, and displays polygenic and complex features. This study aimed to identify the transcriptomic FA profile in the longissimus thoracis muscle in Nellore beef cattle finished in feedlot. Forty-four young bulls were sampled to assess the beef FA profile by considering 14 phenotypes and including differentially expressed genes (DEG), co-expressed (COE), and differentially co-expressed genes (DCO) analyses. All samples (n = 44) were used for COE analysis, whereas 30 samples with extreme phenotypes for the beef FA profile were used for DEG and DCO. A total of 912 DEG were identified, and the polyunsaturated (n = 563) and unsaturated ω-3 (n = 346) FA sums groups were the most frequently observed. The COE analyses identified three modules, of which the blue module (n = 1776) was correlated with eight of 14 FA phenotypes. Also, 759 DCO genes were listed, and the oleic acid (n = 358) and monounsaturated fatty acids sum (n = 120) were the most frequent. Furthermore, 243 and 13, 319 and seven, and 173 and 12 gene ontology terms and Kyoto Encyclopedia of Genes and Genomes pathways were enriched respectively for the DEG, COE, and DCO analyses. Combining the results, we highlight the unexplored GIPC2, ASB5, and PPP5C genes in cattle. Besides LIPE and INSIG2 genes in COE modules, the ACSL3, ECI1, DECR2, FITM1, and SDHB genes were signaled in at least two analyses. These findings contribute to understand the genetic mechanisms underlying the beef FA profile in Nellore beef cattle finished in feedlot.
Collapse
Affiliation(s)
- Gustavo Pimenta Schettini
- School of Agricultural and Veterinary Sciences (FCAV), São Paulo State University (UNESP), Jaboticabal, Brazil
| | - Elisa Peripolli
- School of Veterinary Medicine and Animal Science (FMVZ), University of São Paulo (USP), Pirassununga, Brazil
| | - Pâmela Almeida Alexandre
- Commonwealth Scientific and Industrial Research Organization (CSIRO), Agriculture & Food, Birsbane, Queensland, Australia
| | | | - João Barbosa da Silva Neto
- School of Agricultural and Veterinary Sciences (FCAV), São Paulo State University (UNESP), Jaboticabal, Brazil
| | | | - Lúcia Galvão de Albuquerque
- School of Agricultural and Veterinary Sciences (FCAV), São Paulo State University (UNESP), Jaboticabal, Brazil
| | - Rogério Abdallah Curi
- School of Veterinary Medicine and Animal Science (FMVZ), São Paulo State University (UNESP), Botucatu, Brazil
| | - Fernando Baldi
- School of Agricultural and Veterinary Sciences (FCAV), São Paulo State University (UNESP), Jaboticabal, Brazil
| |
Collapse
|
39
|
Zheng X, Ho QWC, Chua M, Stelmashenko O, Yeo XY, Muralidharan S, Torta F, Chew EGY, Lian MM, Foo JN, Jung S, Wong SH, Tan NS, Tong N, Rutter GA, Wenk MR, Silver DL, Berggren PO, Ali Y. Destabilization of β Cell FIT2 by saturated fatty acids alter lipid droplet numbers and contribute to ER stress and diabetes. Proc Natl Acad Sci U S A 2022; 119:e2113074119. [PMID: 35254894 PMCID: PMC8931238 DOI: 10.1073/pnas.2113074119] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 01/29/2022] [Indexed: 02/05/2023] Open
Abstract
SignificanceWith obesity on the rise, there is a growing appreciation for intracellular lipid droplet (LD) regulation. Here, we show how saturated fatty acids (SFAs) reduce fat storage-inducing transmembrane protein 2 (FIT2)-facilitated, pancreatic β cell LD biogenesis, which in turn induces β cell dysfunction and death, leading to diabetes. This mechanism involves direct acylation of FIT2 cysteine residues, which then marks the FIT2 protein for endoplasmic reticulum (ER)-associated degradation. Loss of β cell FIT2 and LDs reduces insulin secretion, increases intracellular ceramides, stimulates ER stress, and exacerbates diet-induced diabetes in mice. While palmitate and stearate degrade FIT2, unsaturated fatty acids such as palmitoleate and oleate do not, results of which extend to nutrition and diabetes.
Collapse
Affiliation(s)
- Xiaofeng Zheng
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
- Singapore Eye Research Institute, Singapore General Hospital, S168751, Singapore
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Qing Wei Calvin Ho
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
| | - Minni Chua
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
| | - Olga Stelmashenko
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
- Singapore Eye Research Institute, Singapore General Hospital, S168751, Singapore
| | - Xin Yi Yeo
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, S138667, Singapore
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, S119228, Singapore
| | - Sneha Muralidharan
- Singapore Lipidomics Incubator, Department of Medicine, National University of Singapore, S117456, Singapore
| | - Federico Torta
- Singapore Lipidomics Incubator, Department of Biochemistry, Life Sciences Institute and Yong Loo Lin School of Medicine, National University of Singapore, S117456, Singapore
| | - Elaine Guo Yan Chew
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
- Human Genetics, A*STAR, Genome Institute of Singapore, S138672, Singapore
| | - Michelle Mulan Lian
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
- Human Genetics, A*STAR, Genome Institute of Singapore, S138672, Singapore
| | - Jia Nee Foo
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
- Human Genetics, A*STAR, Genome Institute of Singapore, S138672, Singapore
| | - Sangyong Jung
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, S138667, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, S117593, Singapore
| | - Sunny Hei Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
- School of Biological Sciences, Nanyang Technological University Singapore, S637551, Singapore
| | - Nanwei Tong
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Guy A. Rutter
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology, and Metabolism, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London SW7 2AZ, United Kingdom
- Le Centre de recherche du Centre hospitalier de l’Université de Montréal (CR-CHUM), University of Montréal, Montréal, QC H2X 0A9, Canada
| | - Markus R. Wenk
- Singapore Lipidomics Incubator, Department of Biochemistry, Life Sciences Institute and Yong Loo Lin School of Medicine, National University of Singapore, S117456, Singapore
| | - David L. Silver
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke–National University of Singapore Graduate Medical School, S169857, Singapore
| | - Per-Olof Berggren
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
- Singapore Eye Research Institute, Singapore General Hospital, S168751, Singapore
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, People’s Republic of China
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Yusuf Ali
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, S308232, Singapore
- Singapore Eye Research Institute, Singapore General Hospital, S168751, Singapore
| |
Collapse
|
40
|
Parra-Peralbo E, Talamillo A, Barrio R. Origin and Development of the Adipose Tissue, a Key Organ in Physiology and Disease. Front Cell Dev Biol 2022; 9:786129. [PMID: 34993199 PMCID: PMC8724577 DOI: 10.3389/fcell.2021.786129] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/01/2021] [Indexed: 12/17/2022] Open
Abstract
Adipose tissue is a dynamic organ, well known for its function in energy storage and mobilization according to nutrient availability and body needs, in charge of keeping the energetic balance of the organism. During the last decades, adipose tissue has emerged as the largest endocrine organ in the human body, being able to secrete hormones as well as inflammatory molecules and having an important impact in multiple processes such as adipogenesis, metabolism and chronic inflammation. However, the cellular progenitors, development, homeostasis and metabolism of the different types of adipose tissue are not fully known. During the last decade, Drosophila melanogaster has demonstrated to be an excellent model to tackle some of the open questions in the field of metabolism and development of endocrine/metabolic organs. Discoveries ranged from new hormones regulating obesity to subcellular mechanisms that regulate lipogenesis and lipolysis. Here, we review the available evidences on the development, types and functions of adipose tissue in Drosophila and identify some gaps for future research. This may help to understand the cellular and molecular mechanism underlying the pathophysiology of this fascinating key tissue, contributing to establish this organ as a therapeutic target.
Collapse
Affiliation(s)
| | - Ana Talamillo
- Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Rosa Barrio
- Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| |
Collapse
|
41
|
Siddiqui S, Glauben R. Fatty Acid Metabolism in Myeloid-Derived Suppressor Cells and Tumor-Associated Macrophages: Key Factor in Cancer Immune Evasion. Cancers (Basel) 2022; 14:250. [PMID: 35008414 PMCID: PMC8750448 DOI: 10.3390/cancers14010250] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 12/16/2021] [Accepted: 12/27/2021] [Indexed: 02/04/2023] Open
Abstract
The tumor microenvironment (TME) comprises various cell types, soluble factors, viz, metabolites or cytokines, which together play in promoting tumor metastasis. Tumor infiltrating immune cells play an important role against cancer, and metabolic switching in immune cells has been shown to affect activation, differentiation, and polarization from tumor suppressive into immune suppressive phenotypes. Macrophages represent one of the major immune infiltrates into TME. Blood monocyte-derived macrophages and myeloid derived suppressor cells (MDSCs) infiltrating into the TME potentiate hostile tumor progression by polarizing into immunosuppressive tumor-associated macrophages (TAMs). Recent studies in the field of immunometabolism focus on metabolic reprogramming at the TME in polarizing tumor-associated macrophages (TAMs). Lipid droplets (LD), detected in almost every eukaryotic cell type, represent the major source for intra-cellular fatty acids. Previously, LDs were mainly described as storage sites for fatty acids. However, LDs are now recognized to play an integral role in cellular signaling and consequently in inflammation and metabolism-mediated phenotypical changes in immune cells. In recent years, the role of LD dependent metabolism in macrophage functionality and phenotype has been being investigated. In this review article, we discuss fatty acids stored in LDs, their role in modulating metabolism of tumor-infiltrating immune cells and, therefore, in shaping the cancer progression.
Collapse
Affiliation(s)
| | - Rainer Glauben
- Medical Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany;
| |
Collapse
|
42
|
Hello from the other side: Membrane contact of lipid droplets with other organelles and subsequent functional implications. Prog Lipid Res 2021; 85:101141. [PMID: 34793861 DOI: 10.1016/j.plipres.2021.101141] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/10/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023]
Abstract
Lipid droplets (LDs) are ubiquitous organelles that play crucial roles in response to physiological and environmental cues. The identification of several neutral lipid synthesizing and regulatory protein complexes have propelled significant advance on the mechanisms of LD biogenesis in the endoplasmic reticulum (ER). Increasing evidence suggests that distinct proteins and regulatory factors, which localize to membrane contact sites (MCS), are involved not only in interorganellar lipid exchange and transport, but also function in other important cellular processes, including autophagy, mitochondrial dynamics and inheritance, ion signaling and inter-regulation of these MCS. More and more tethers and molecular determinants are associated to MCS and to a diversity of cellular and pathophysiological processes, demonstrating the dynamics and importance of these junctions in health and disease. The conjugation of lipids with proteins in supramolecular complexes is known to be paramount for many biological processes, namely membrane biosynthesis, cell homeostasis, regulation of organelle division and biogenesis, and cell growth. Ultimately, this physical organization allows the contact sites to function as crucial metabolic hubs that control the occurrence of chemical reactions. This leads to biochemical and metabolite compartmentalization for the purposes of energetic efficiency and cellular homeostasis. In this review, we will focus on the structural and functional aspects of LD-organelle interactions and how they ensure signaling exchange and metabolites transfer between organelles.
Collapse
|
43
|
Nuclear CoRepressors, NCOR1 and SMRT, are required for maintaining systemic metabolic homeostasis. Mol Metab 2021; 53:101315. [PMID: 34390859 PMCID: PMC8429965 DOI: 10.1016/j.molmet.2021.101315] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/20/2021] [Accepted: 08/09/2021] [Indexed: 02/07/2023] Open
Abstract
Objective The nuclear receptor corepressor 1 (NCOR1) and the silencing mediator of retinoic acid and thyroid hormone (SMRT, also known as NCOR2) play critical and specific roles in nuclear receptor action. NCOR1, both in vitro and in vivo specifically regulates thyroid hormone (TH) action in the context of individual organs such as the liver, and systemically in the context of the hypothalamic-pituitary-thyroid (HPT) axis. In contrast, selective deletion of SMRT in the liver or globally has shown that it plays very little role in TH signaling. However, both NCOR1 and SMRT have some overlapping roles in hepatic metabolism and lipogenesis. Here, we determine the roles of NCOR1 and SMRT in global physiologic function and find if SMRT could play a compensatory role in the regulation of TH action, globally. Methods We used a postnatal deletion strategy to disrupt both NCOR1 and SMRT together in all tissues at 8–9 weeks of age in male and female mice. This was performed using a tamoxifen-inducible Cre recombinase (UBC-Cre-ERT2) to KO (knockout) NCOR1, SMRT, or NCOR1 and SMRT together. We used the same strategy to KO HDAC3 in male and female mice of the same age. Metabolic parameters, gene expression, and thyroid function tests were analyzed. Results Surprisingly, adult mice that acquired NCOR1 and SMRT deletion rapidly became hypoglycemic and hypothermic and perished within ten days of deletion of both corepressors. Postnatal deletion of either NCOR1 or SMRT had no impact on mortality. NCOR1/SMRT KO mice rapidly developed hepatosteatosis and mild elevations in liver function tests. Additionally, alterations in lipogenesis, beta oxidation, along with hepatic triglyceride and glycogen levels suggested defects in hepatic metabolism. The intestinal function was intact in the NCOR1/SMRT knockout (KO) mice. The KO of HDAC3 resulted in a distinct phenotype from the NCOR1/SMRT KO mice, whereas none of the HDAC3 KO mice succumbed after tamoxifen injection. Conclusions The KO of NCOR1 and SMRT rapidly leads to significant metabolic abnormalities that do not survive – including hypoglycemia, hypothermia, and weight loss. Hepatosteatosis rapidly developed along with alterations in hepatic metabolism suggesting a contribution to the dramatic phenotype from liver injury. Glucose production and absorption were intact in NCOR1/SMRT KO mice, demonstrating a multifactorial process leading to their demise. HDAC3 KO mice have a distinct phenotype from the NCOR1/SMRT KO mice—which implies that NCOR1/SMRT together regulate a critical pathway that is required for survival in adulthood and is separate from HDAC3. The knockout of corepressors NCoR1 and SMRT is rapidly lethal. Metabolic abnormalities observed include hypoglycemia and hypothermia. Hepatic glucose production and intestinal absorption is intact despite hypoglycemia. The lethal action of NCoR1/SMRT deletion is independent of HDAC3.
Collapse
|
44
|
Rahman MA, Kumar R, Sanchez E, Nazarko TY. Lipid Droplets and Their Autophagic Turnover via the Raft-Like Vacuolar Microdomains. Int J Mol Sci 2021; 22:8144. [PMID: 34360917 PMCID: PMC8348048 DOI: 10.3390/ijms22158144] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/26/2021] [Accepted: 07/26/2021] [Indexed: 01/01/2023] Open
Abstract
Although once perceived as inert structures that merely serve for lipid storage, lipid droplets (LDs) have proven to be the dynamic organelles that hold many cellular functions. The LDs' basic structure of a hydrophobic core consisting of neutral lipids and enclosed in a phospholipid monolayer allows for quick lipid accessibility for intracellular energy and membrane production. Whereas formed at the peripheral and perinuclear endoplasmic reticulum, LDs are degraded either in the cytosol by lipolysis or in the vacuoles/lysosomes by autophagy. Autophagy is a regulated breakdown of dysfunctional, damaged, or surplus cellular components. The selective autophagy of LDs is called lipophagy. Here, we review LDs and their degradation by lipophagy in yeast, which proceeds via the micrometer-scale raft-like lipid domains in the vacuolar membrane. These vacuolar microdomains form during nutrient deprivation and facilitate internalization of LDs via the vacuolar membrane invagination and scission. The resultant intra-vacuolar autophagic bodies with LDs inside are broken down by vacuolar lipases and proteases. This type of lipophagy is called microlipophagy as it resembles microautophagy, the type of autophagy when substrates are sequestered right at the surface of a lytic compartment. Yeast microlipophagy via the raft-like vacuolar microdomains is a great model system to study the role of lipid domains in microautophagic pathways.
Collapse
Affiliation(s)
- Muhammad Arifur Rahman
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (M.A.R.); (E.S.)
| | - Ravinder Kumar
- Department of Obstetrics, Gynecology and Reproductive Science, University of California, San Francisco, CA 94143, USA;
| | - Enrique Sanchez
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (M.A.R.); (E.S.)
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Taras Y. Nazarko
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (M.A.R.); (E.S.)
| |
Collapse
|
45
|
Brink JTR, Fourie R, Sebolai O, Albertyn J, Pohl CH. The role of lipid droplets in microbial pathogenesis. J Med Microbiol 2021; 70. [PMID: 34184983 DOI: 10.1099/jmm.0.001383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The nonpolar lipids present in cells are mainly triacylglycerols and steryl esters. When cells are provided with an abundance of nutrients, these storage lipids accumulate. As large quantities of nonpolar lipids cannot be integrated into membranes, they are isolated from the cytosolic environment in lipid droplets. As specialized, inducible cytoplasmic organelles, lipid droplets have functions beyond the regulation of lipid metabolism, in cell signalling and activation, membrane trafficking and control of inflammatory mediator synthesis and secretion. Pathogens, including fungi, viruses, parasites, or intracellular bacteria can induce and may benefit from lipid droplets in infected cells. Here we review biogenesis of lipid droplets as well as the role of lipid droplets in the pathogenesis of selected viruses, bacteria, protists and yeasts.
Collapse
Affiliation(s)
- Jacobus T R Brink
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Ruan Fourie
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Olihile Sebolai
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Jacobus Albertyn
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Carolina H Pohl
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| |
Collapse
|
46
|
Bagchi DP, MacDougald OA. Wnt Signaling: From Mesenchymal Cell Fate to Lipogenesis and Other Mature Adipocyte Functions. Diabetes 2021; 70:1419-1430. [PMID: 34155042 PMCID: PMC8336005 DOI: 10.2337/dbi20-0015] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/20/2021] [Indexed: 12/14/2022]
Abstract
Wnt signaling is an ancient and evolutionarily conserved pathway with fundamental roles in the development of adipose tissues. Roles of this pathway in mesenchymal stem cell fate determination and differentiation have been extensively studied. Indeed, canonical Wnt signaling is a significant endogenous inhibitor of adipogenesis and promoter of other cell fates, including osteogenesis, chondrogenesis, and myogenesis. However, emerging genetic evidence in both humans and mice suggests central roles for Wnt signaling in body fat distribution, obesity, and metabolic dysfunction. Herein, we highlight recent studies that have begun to unravel the contributions of various Wnt pathway members to critical adipocyte functions, including carbohydrate and lipid metabolism. We further explore compelling evidence of complex and coordinated interactions between adipocytes and other cell types within adipose tissues, including stromal, immune, and endothelial cells. Given the evolutionary conservation and ubiquitous cellular distribution of this pathway, uncovering the contributions of Wnt signaling to cell metabolism has exciting implications for therapeutic intervention in widespread pathologic states, including obesity, diabetes, and cancers.
Collapse
Affiliation(s)
- Devika P Bagchi
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI
| | - Ormond A MacDougald
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
47
|
Kellermann M, Scharte F, Hensel M. Manipulation of Host Cell Organelles by Intracellular Pathogens. Int J Mol Sci 2021; 22:ijms22126484. [PMID: 34204285 PMCID: PMC8235465 DOI: 10.3390/ijms22126484] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/13/2022] Open
Abstract
Pathogenic intracellular bacteria, parasites and viruses have evolved sophisticated mechanisms to manipulate mammalian host cells to serve as niches for persistence and proliferation. The intracellular lifestyles of pathogens involve the manipulation of membrane-bound organellar compartments of host cells. In this review, we described how normal structural organization and cellular functions of endosomes, endoplasmic reticulum, Golgi apparatus, mitochondria, or lipid droplets are targeted by microbial virulence mechanisms. We focus on the specific interactions of Salmonella, Legionella pneumophila, Rickettsia rickettsii, Chlamydia spp. and Mycobacterium tuberculosis representing intracellular bacterial pathogens, and of Plasmodium spp. and Toxoplasma gondii representing intracellular parasites. The replication strategies of various viruses, i.e., Influenza A virus, Poliovirus, Brome mosaic virus, Epstein-Barr Virus, Hepatitis C virus, severe acute respiratory syndrome virus (SARS), Dengue virus, Zika virus, and others are presented with focus on the specific manipulation of the organelle compartments. We compare the specific features of intracellular lifestyle and replication cycles, and highlight the communalities in mechanisms of manipulation deployed.
Collapse
Affiliation(s)
- Malte Kellermann
- Abt. Mikrobiologie, Fachbereich Biologie/Chemie, Barbarastr 11, Universität Osnabrück, 49076 Osnabrück, Germany; (M.K.); (F.S.)
| | - Felix Scharte
- Abt. Mikrobiologie, Fachbereich Biologie/Chemie, Barbarastr 11, Universität Osnabrück, 49076 Osnabrück, Germany; (M.K.); (F.S.)
| | - Michael Hensel
- Abt. Mikrobiologie, Fachbereich Biologie/Chemie, Barbarastr 11, Universität Osnabrück, 49076 Osnabrück, Germany; (M.K.); (F.S.)
- CellNanOs–Center of Cellular Nanoanalytics Osnabrück, Universität Osnabrück, Barbarastr 11, 49076 Osnabrück, Germany
- Correspondence: ; Tel.: +49-(0)-541-969-3940
| |
Collapse
|
48
|
Khaddaj R, Mari M, Cottier S, Reggiori F, Schneiter R. The surface of lipid droplets constitutes a barrier for endoplasmic reticulum-resident integral membrane proteins. J Cell Sci 2021; 135:268334. [PMID: 34028531 DOI: 10.1242/jcs.256206] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 04/14/2021] [Indexed: 12/12/2022] Open
Abstract
Lipid droplets (LDs) are globular subcellular structures that store neutral lipids. LDs are closely associated with the endoplasmic reticulum (ER) and are limited by a phospholipid monolayer harboring a specific set of proteins. Most of these proteins associate with LDs through either an amphipathic helix or a membrane-embedded hairpin motif. Here, we address the question of whether integral membrane proteins can localize to the surface of LDs. To test this, we fused perilipin 3 (PLIN3), a mammalian LD-targeted protein, to ER-resident proteins. The resulting fusion proteins localized to the periphery of LDs in both yeast and mammalian cells. This peripheral LD localization of the fusion proteins, however, was due to a redistribution of the ER around LDs, as revealed by bimolecular fluorescence complementation between ER- and LD-localized partners. A LD-tethering function of PLIN3-containing membrane proteins was confirmed by fusing PLIN3 to the cytoplasmic domain of an outer mitochondrial membrane protein, OM14. Expression of OM14-PLIN3 induced a close apposition between LDs and mitochondria. These data indicate that the ER-LD junction constitutes a barrier for ER-resident integral membrane proteins.
Collapse
Affiliation(s)
- Rasha Khaddaj
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Muriel Mari
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Stéphanie Cottier
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Fulvio Reggiori
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Roger Schneiter
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| |
Collapse
|
49
|
Chen F, Yan B, Ren J, Lyu R, Wu Y, Guo Y, Li D, Zhang H, Hu J. FIT2 organizes lipid droplet biogenesis with ER tubule-forming proteins and septins. J Cell Biol 2021; 220:211999. [PMID: 33861319 PMCID: PMC8056755 DOI: 10.1083/jcb.201907183] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/29/2020] [Accepted: 02/09/2021] [Indexed: 12/20/2022] Open
Abstract
Lipid droplets (LDs) are critical for lipid storage and energy metabolism. LDs form in the endoplasmic reticulum (ER). However, the molecular basis for LD biogenesis remains elusive. Here, we show that fat storage–inducing transmembrane protein 2 (FIT2) interacts with ER tubule-forming proteins Rtn4 and REEP5. The association is mainly transmembrane domain based and stimulated by oleic acid. Depletion of ER tubule-forming proteins decreases the number and size of LDs in cells and Caenorhabditis elegans, mimicking loss of FIT2. Through cytosolic loops, FIT2 binds to cytoskeletal protein septin 7, an interaction that is also required for normal LD biogenesis. Depletion of ER tubule-forming proteins or septins delays nascent LD formation. In addition, FIT2-interacting proteins are up-regulated during adipocyte differentiation, and ER tubule-forming proteins, septin 7, and FIT2 are transiently enriched at LD formation sites. Thus, FIT2-mediated nascent LD biogenesis is facilitated by ER tubule-forming proteins and septins.
Collapse
Affiliation(s)
- Fang Chen
- National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Bing Yan
- National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jie Ren
- National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Rui Lyu
- National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yanfang Wu
- National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yuting Guo
- National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Dong Li
- National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Hong Zhang
- National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Junjie Hu
- National Laboratory of Biomacromolecules, Chinese Academy of Sciences Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
50
|
Choudhary V, Schneiter R. A Unique Junctional Interface at Contact Sites Between the Endoplasmic Reticulum and Lipid Droplets. Front Cell Dev Biol 2021; 9:650186. [PMID: 33898445 PMCID: PMC8060488 DOI: 10.3389/fcell.2021.650186] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/09/2021] [Indexed: 12/19/2022] Open
Abstract
Lipid droplets (LDs) constitute compartments dedicated to the storage of metabolic energy in the form of neutral lipids. LDs originate from the endoplasmic reticulum (ER) with which they maintain close contact throughout their life cycle. These ER-LD junctions facilitate the exchange of both proteins and lipids between these two compartments. In recent years, proteins that are important for the proper formation of LDs and localize to ER-LD junctions have been identified. This junction is unique as it is generally believed to invoke a transition from the ER bilayer membrane to a lipid monolayer that delineates LDs. Proper formation of this junction requires the ordered assembly of proteins and lipids at specialized ER subdomains. Without such a well-ordered assembly of LD biogenesis factors, neutral lipids are synthesized throughout the ER membrane, resulting in the formation of aberrant LDs. Such ectopically formed LDs impact ER and lipid homeostasis, resulting in different types of lipid storage diseases. In response to starvation, the ER-LD junction recruits factors that tether the vacuole to these junctions to facilitate LD degradation. In addition, LDs maintain close contacts with peroxisomes and mitochondria for metabolic channeling of the released fatty acids toward beta-oxidation. In this review, we discuss the function of different components that ensure proper functioning of LD contact sites, their role in lipogenesis and lipolysis, and their relation to lipid storage diseases.
Collapse
Affiliation(s)
- Vineet Choudhary
- Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Roger Schneiter
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|