1
|
Park J, Na CS. Weighted single-step genome-wide association study to reveal new candidate genes for productive traits of Landrace pig in Korea. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2024; 66:702-716. [PMID: 39165735 PMCID: PMC11331376 DOI: 10.5187/jast.2024.e104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/12/2023] [Accepted: 10/04/2023] [Indexed: 08/22/2024]
Abstract
The objective of this study was to identify genomic regions and candidate genes associated with productive traits using a total of 37,099 productive records and 6,683 single nucleotide polymorphism (SNP) data obtained from five Great-Grand-Parents (GGP) farms in Landrace. The estimated of heritabilities for days to 105 kg (AGE), average daily gain (ADG), backfat thickness (BF), and eye muscle area (EMA) were 0.49, 0.49, 0.56, and 0.23, respectively. We identified a genetic window that explained 2.05%-2.34% for each trait of the total genetic variance. We observed a clear partitioning of the four traits into two groups, and the most significant genomic region for AGE and ADG were located on the Sus scrofa chromosome (SSC) 1, while BF and EMA were located on SSC 2. We conducted Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), which revealed results in three biological processes, four cellular component, three molecular function, and six KEGG pathway. Significant SNPs can be used as markers for quantitative trait loci (QTL) investigation and genomic selection (GS) for productive traits in Landrace pig.
Collapse
Affiliation(s)
- Jun Park
- Department of Animal Biotechnology,
Jeonbuk National University, Jeonju 54896, Korea
| | - Chong-Sam Na
- Department of Animal Biotechnology,
Jeonbuk National University, Jeonju 54896, Korea
| |
Collapse
|
2
|
Kobayashi Y, Eguchi A, Imami K, Tempaku M, Izuoka K, Takase T, Kainuma K, Nagao M, Furuta N, Iwasa M, Nakagawa H, Fujisawa T, Togashi K. Circulating extracellular vesicles are associated with pathophysiological condition including metabolic syndrome-related dysmetabolism in children and adolescents with obesity. J Mol Med (Berl) 2024; 102:23-38. [PMID: 37874387 DOI: 10.1007/s00109-023-02386-5] [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: 03/20/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/25/2023]
Abstract
Obesity of children and adolescents (OCA) is often accompanied by metabolic syndrome (MetS), which often leads to adult obesity and subsequent complications, yet the entire pathophysiological response is not fully understood. The number and composition of circulating extracellular vesicles (EV) reflect overall patient condition; therefore, we investigated the pathophysiological condition of OCA, including MetS-associated dysmetabolism, using circulating EVs. In total, 107 children and adolescents with or without obesity (boys, n = 69; girls, n = 38; median age, 10 years) were enrolled. Circulating EV number and EV protein composition were assessed via flow cytometry and liquid chromatography tandem-mass spectrometry, respectively. In a multivariate analysis, relative body weight (standardized partial regression coefficient (SPRC) 0.469, P = 0.012) and serum triglyceride level (SPRC 0.548, P < 0.001) were detected as independent parameters correlating with circulating EV number. Proteomic analysis identified 31 upregulated and 45 downregulated EV proteins in OCA. Gene ontology analysis revealed upregulated proteins to be involved in various biological processes, including intracellular protein transport, protein folding, stress response, leukocyte activation, innate immune response, and platelet degranulation, which can modulate lipid and glucose metabolism, skeletal and cardiac muscle development, inflammation, immune response, carcinogenesis, and cancer progression. Notably, several identified EV proteins are involved in neuro-development, neurotransmitter release, and neuro-protective agents in OCA. Circulating EVs were derived from adipocytes, hepatocytes, B cell lymphocytes, and neurons. Circulating EV number is significantly associated with MetS-related dysmetabolism and the EV protein cargo carries a special "signature" that reflects the alteration of various biological processes under the pathophysiological condition of OCA. KEY MESSAGES: Circulating EV number correlates with physical and laboratory parameters for obesity in children and adolescents. Relative body weight and triglyceride are independent factors for increased circulating EVs. EV composition is significantly changed in obesity of children and adolescents. Identified EV composition changes associated with obesity and involves in metabolism, immune response, and cancer progression. Circulating EVs are partially derived from adipocyte, hepatocytes, B cells, and neurons.
Collapse
Affiliation(s)
- Yoshinao Kobayashi
- Center for Physical and Mental Health, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Akiko Eguchi
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan.
- JST, PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan.
- Biobank Center, Mie University Hospital, Tsu, Mie, 514-8507, Japan.
| | - Koshi Imami
- JST, PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Mina Tempaku
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Kiyora Izuoka
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Takafumi Takase
- Department of Pediatrics, National Hospital Organization Mie National Hospital, Tsu, Mie, 514-0125, Japan
| | - Keigo Kainuma
- Department of Pediatrics, National Hospital Organization Mie National Hospital, Tsu, Mie, 514-0125, Japan
| | - Mizuho Nagao
- Department of Pediatrics, National Hospital Organization Mie National Hospital, Tsu, Mie, 514-0125, Japan
| | - Noriko Furuta
- Center for Physical and Mental Health, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan
| | - Motoh Iwasa
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Hayato Nakagawa
- Department of Gastroenterology and Hepatology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Takao Fujisawa
- Department of Pediatrics, National Hospital Organization Mie National Hospital, Tsu, Mie, 514-0125, Japan
| | - Kenji Togashi
- Department of Health and Physical Education, Faculty of Education, Mie University, Tsu, Mie, 514-8507, Japan
| |
Collapse
|
3
|
Zhang L, Zhang W, Xiao C, Wu X, Cui H, Yan P, Yang C, Tang M, Wang Y, Chen L, Liu Y, Zou Y, Alfredsson L, Klareskog L, Yang Y, Yao Y, Li J, Liu Z, Yang C, Jiang X, Zhang B. Using human genetics to understand the epidemiological association between obesity, serum urate, and gout. Rheumatology (Oxford) 2023; 62:3280-3290. [PMID: 36734534 DOI: 10.1093/rheumatology/kead054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/31/2022] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVES We aimed to clarify the genetic overlaps underlying obesity-related traits, serum urate, and gout. METHODS We conducted a comprehensive genome-wide cross-trait analysis to identify genetic correlation, pleiotropic loci, and causal relationships between obesity (the exposure variable), gout (the primary outcome) and serum urate (the secondary outcome). Summary statistics were collected from the hitherto largest genome-wide association studies conducted for BMI (N = 806 834), waist-to-hip ratio (WHR; N = 697 734), WHR adjusted for BMI (WHRadjBMI; N = 694 649), serum urate (N = 288 649), and gout (Ncases = 13 179 and Ncontrols = 750 634). RESULTS Positive overall genetic correlations were observed for BMI (rg = 0.27, P = 6.62 × 10-7), WHR (rg = 0.22, P = 6.26 × 10-7) and WHRadjBMI (rg = 0.07, P = 6.08 × 10-3) with gout. Partitioning the whole genome into 1703 LD (linkage disequilibrium)-independent regions, a significant local signal at 4q22 was identified for BMI and gout. The global and local shared genetic basis was further strengthened by the multiple pleiotropic loci identified in the cross-phenotype association study, multiple shared gene-tissue pairs observed by Transcriptome-wide association studies, as well as causal relationships demonstrated by Mendelian randomization [BMI-gout: OR (odds ratio) = 1.66, 95% CI = 1.45, 1.88; WHR-gout: OR = 1.57, 95% CI = 1.37, 1.81]. Replacing the binary disease status of gout with its latent pathological measure, serum urate, a similar pattern of correlation, pleiotropy and causality was observed with even more pronounced magnitude and significance. CONCLUSION Our comprehensive genome-wide cross-trait analysis demonstrates a shared genetic basis and pleiotropic loci, as well as a causal relationship between obesity, serum urate, and gout, highlighting an intrinsic link underlying these complex traits.
Collapse
Affiliation(s)
- Li Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Wenqiang Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chenghan Xiao
- Department of Maternal, Child and Adolescent Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xueyao Wu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Huijie Cui
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Peijing Yan
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chao Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Mingshuang Tang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yutong Wang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Lin Chen
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yunjie Liu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yanqiu Zou
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Lars Alfredsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Lars Klareskog
- Division of Rheumatology, Department of Medicine and Center for Molecular Medicine, Karolinska Institutet at Karolinska University Hospital (Solna), Stockholm, Sweden
| | - Yanfang Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yuqin Yao
- Department of Occupational and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jiayuan Li
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Zhenmi Liu
- Department of Maternal, Child and Adolescent Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chunxia Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xia Jiang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Ben Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Epidemiology and Biostatistics, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
- Department of Occupational and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Maharjan Y, Dutta RK, Son J, Wei X, Park C, Kwon HM, Park R. Intracellular cholesterol transport inhibition Impairs autophagy flux by decreasing autophagosome-lysosome fusion. Cell Commun Signal 2022; 20:189. [PMID: 36434621 PMCID: PMC9701069 DOI: 10.1186/s12964-022-00942-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/17/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Autophagy is an intracellular degradation process crucial for homeostasis. During autophagy, a double-membrane autophagosome fuses with lysosome through SNARE machinery STX17 to form autolysosome for degradation of damaged organelle. Whereas defective autophagy enhances cholesterol accumulation in the lysosome and impaired autophagic flux that results Niemann-Pick type C1 (NPC1) disease. However, exact interconnection between NPC1 and autophagic flux remain obscure due to the existence of controversial reports. RESULTS This study aimed at a comparison of the effects of three autophagic inhibitor drugs, including chloroquine, U18666A, and bafilomycin A1, on the intracellular cholesterol transport and autophagy flux. Chloroquine, an autophagic flux inhibitor; U1866A, a NPC1 inhibitor, and bafilomycin A, a lysosomotropic agent are well known to inhibit autophagy by different mechanism. Here we showed that treatment with U1866A and bafilomycin A induces lysosomal cholesterol accumulation that prevented autophagic flux by decreasing autophagosome-lysosome fusion. We also demonstrated that accumulation of cholesterol within the lysosome did not affect lysosomal pH. Although the clearance of accumulated cholesterol by cyclodextrin restored the defective autophagosome-lysosome fusion, the autophagy flux restoration was possible only when lysosomal acidification was not altered. In addition, a failure of STX17 trafficking to autophagosomes plays a key role in prevention of autophagy flux caused by intracellular cholesterol transport inhibitors. CONCLUSIONS Our data provide a new insight that the impaired autophagy flux does not necessarily result in lysosomal cholesterol accumulation even though it prevents autophagosome-lysosome fusion. Video abstract.
Collapse
Affiliation(s)
- Yunash Maharjan
- grid.61221.360000 0001 1033 9831Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005 Republic of Korea ,grid.224260.00000 0004 0458 8737School of Dentistry, Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, VA USA ,grid.224260.00000 0004 0458 8737Present Address: Department of Oral and Craniofacial Molecular Biology, School of Dentistry, Virginia Commonwealth University, Richmond, VA USA
| | - Raghbendra Kumar Dutta
- grid.61221.360000 0001 1033 9831Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005 Republic of Korea
| | - Jinbae Son
- grid.61221.360000 0001 1033 9831Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005 Republic of Korea
| | - Xiaofan Wei
- grid.61221.360000 0001 1033 9831Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005 Republic of Korea
| | - Channy Park
- grid.61221.360000 0001 1033 9831Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005 Republic of Korea
| | - Hyug Moo Kwon
- grid.42687.3f0000 0004 0381 814XSchool of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Raekil Park
- grid.61221.360000 0001 1033 9831Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005 Republic of Korea
| |
Collapse
|
5
|
Woo JH, Park SJ, Park SM, Joe E, Jou I. Interleukin‐6 signaling requires EHD1‐mediated alteration of membrane rafts. FEBS J 2022; 289:5914-5932. [DOI: 10.1111/febs.16458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/25/2022] [Accepted: 04/12/2022] [Indexed: 01/09/2023]
Affiliation(s)
- Joo Hong Woo
- Inflamm‐aging Translational Research Center Ajou University School of Medicine Suwon Korea
| | - Soo Jung Park
- Inflamm‐aging Translational Research Center Ajou University School of Medicine Suwon Korea
| | - Sang Myun Park
- Department of Pharmacology Ajou University School of Medicine Suwon Korea
- Center for Convergence Research of Neurological Disorders Ajou University School of Medicine Suwon Korea
| | - Eun‐hye Joe
- Department of Pharmacology Ajou University School of Medicine Suwon Korea
- Center for Convergence Research of Neurological Disorders Ajou University School of Medicine Suwon Korea
| | - Ilo Jou
- Inflamm‐aging Translational Research Center Ajou University School of Medicine Suwon Korea
- Department of Pharmacology Ajou University School of Medicine Suwon Korea
| |
Collapse
|
6
|
Abstract
Interorganellar cross talk is often mediated by membrane contact sites (MCSs), which are zones where participating membranes come within 30 nm of one another. MCSs have been found in organelles, including the endoplasmic reticulum, Golgi bodies, endosomes, and mitochondria. Despite its seeming ubiquity, reports of MCS involving mitochondrion-related organelles (MROs) present in a few anaerobic parasitic protozoa remain lacking. Entamoeba histolytica, the etiological agent of amoebiasis, possesses an MRO called the mitosome. We previously discovered several Entamoeba-specific transmembrane mitosomal proteins (ETMPs) from in silico and cell-biological analyses. One of them, ETMP1 (EHI_175060), was predicted to have one transmembrane domain and two coiled-coil regions and was demonstrated to be mitosome membrane integrated based on carbonate fractionation and immunoelectron microscopy (IEM) data. Immunoprecipitation analysis detected a candidate interacting partner, EH domain-containing protein (EHD1; EHI_105270). We expressed hemagglutinin (HA)-tagged EHD1 in E. histolytica, and subsequent immunofluorescence and IEM data indicated an unprecedented MCS between the mitosome and the endosome. Live imaging of a green fluorescent protein (GFP)-EHD1-expressing strain demonstrated that EHD1 is involved in early endosome formation and is observed in MCS between endosomes of various sizes. In vitro assays using recombinant His-EHD1 demonstrated ATPase activity. MCSs are involved in lipid transfer, ion homeostasis, and organelle dynamics. The serendipitous discovery of the ETMP1-interacting partner EHD1 led to the observation of the mitosome-endosome contact site in E. histolytica. It opened a new view of how the relic mitochondria of Entamoeba may likewise be involved in organelle cross talk, a conserved feature of mitochondria and other organelles in general.
Collapse
|
7
|
Yang W, Liu Z, Zhao Q, Du H, Yu J, Wang H, Liu X, Liu H, Jing X, Yang H, Shi G, Zhou L, Liu J. Population Genetic Structure and Selection Signature Analysis of Beijing Black Pig. Front Genet 2022; 13:860669. [PMID: 35401688 PMCID: PMC8987279 DOI: 10.3389/fgene.2022.860669] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/08/2022] [Indexed: 12/04/2022] Open
Abstract
Beijing Black pig is an excellent cultivated black pig breed in China, with desirable body shape, tender meat quality and robust disease resistance. To explore the level of admixture and selection signatures of Beijing Black pigs, a total number of 90 individuals covering nine pig breeds were used in our study, including Beijing Black pig, Large White, Landrace, Duroc, Lantang pig, Luchuan pig, Mashen pig, Huainan pig and Min pig. These animals were resequenced with 18.19 folds mapped read depth on average. Generally, we found that Beijing Black pig was genetically closer to commercial pig breeds by population genetic structure and genetic diversity analysis, and was also affected by Chinese domestic breeds Huainan pig and Min pig. These results are consistent with the cross-breeding history of Beijing Black pig. Selection signal detections were performed on three pig breeds, Beijing Black pig, Duroc and Large White, using three complementary methods (FST, θπ, and XP-EHH). In total, 1,167 significant selected regions and 392 candidate genes were identified. Functional annotations were enriched to pathways related to immune processes and meat and lipid metabolism. Finally, potential candidate genes, influencing meat quality (GPHA2, EHD1, HNF1A, C12orf43, GLTP, TRPV4, MVK, and MMAB), reproduction (PPP2R5B and MAP9), and disease resistance (OASL, ANKRD13A, and GIT2), were further detected by gene annotation analysis. Our results advanced the understanding of the genetic mechanism behind artificial selection of Beijing Black pigs, and provided theoretical basis for the subsequent breeding and genetic research of this breed.
Collapse
Affiliation(s)
- Wenjing Yang
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhen Liu
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qiqi Zhao
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Heng Du
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jian Yu
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Hongwei Wang
- Beijing Heiliu Stockbreeding Technology Co.,Ltd, Beijing, China
| | - Xiance Liu
- Beijing Heiliu Stockbreeding Technology Co.,Ltd, Beijing, China
| | - Hai Liu
- Beijing Heiliu Stockbreeding Technology Co.,Ltd, Beijing, China
| | - Xitao Jing
- Beijing Heiliu Stockbreeding Technology Co.,Ltd, Beijing, China
| | - Hongping Yang
- Beijing Heiliu Stockbreeding Technology Co.,Ltd, Beijing, China
| | - Guohua Shi
- Beijing Heiliu Stockbreeding Technology Co.,Ltd, Beijing, China
| | - Lei Zhou
- College of Animal Science and Technology, China Agricultural University, Beijing, China
- *Correspondence: Lei Zhou, ; Jianfeng Liu,
| | - Jianfeng Liu
- College of Animal Science and Technology, China Agricultural University, Beijing, China
- *Correspondence: Lei Zhou, ; Jianfeng Liu,
| |
Collapse
|
8
|
Maharjan Y, Lee JN, Kwak SA, Dutta RK, Park C, Choe SK, Park R. TMEM135 regulates primary ciliogenesis through modulation of intracellular cholesterol distribution. EMBO Rep 2020; 21:e48901. [PMID: 32157776 PMCID: PMC7202201 DOI: 10.15252/embr.201948901] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 01/31/2020] [Accepted: 02/14/2020] [Indexed: 11/09/2022] Open
Abstract
Recent evidence has linked the lysosomal cholesterol accumulation in Niemann–Pick type C1 with anomalies associated with primary ciliogenesis. Here, we report that perturbed intracellular cholesterol distribution imposed by lysosomal cholesterol accumulation during TMEM135 depletion is closely associated with impaired ciliogenesis. TMEM135 depletion does not affect the formation of the basal body and the ciliary transition zone. TMEM135 depletion severely blunts Rab8 trafficking to the centrioles without affecting the centriolar localization of Rab11 and Rabin8, the upstream regulators of Rab8 activation. Although TMEM135 depletion prevents enhanced IFT20 localization at the centrioles, ciliary vesicle formation is not affected. Furthermore, enhanced IFT20 localization at the centrioles is dependent on Rab8 activation. Supplementation of cholesterol in complex with cyclodextrin rescues Rab8 trafficking to the centrioles and Rab8 activation, thereby recovering primary ciliogenesis in TMEM135‐depleted cells. Taken together, our data suggest that TMEM135 depletion prevents ciliary vesicle elongation, a characteristic of impaired Rab8 function. Our study thus reveals a previously uncharacterized effect of erroneous intracellular cholesterol distribution on impairing Rab8 function and primary ciliogenesis.
Collapse
Affiliation(s)
- Yunash Maharjan
- Department of Biomedical Science & Engineering, Gwangju Institute of Science & Technology, Gwangju, Korea
| | - Joon No Lee
- Department of Biomedical Science & Engineering, Gwangju Institute of Science & Technology, Gwangju, Korea
| | - Seong Ae Kwak
- Department of Microbiology and Center for Metabolic Function Regulation, Wonkwang University School of Medicine, Iksan, Korea
| | - Raghbendra Kumar Dutta
- Department of Biomedical Science & Engineering, Gwangju Institute of Science & Technology, Gwangju, Korea
| | - Channy Park
- Department of Biomedical Science & Engineering, Gwangju Institute of Science & Technology, Gwangju, Korea
| | - Seong-Kyu Choe
- Department of Microbiology and Center for Metabolic Function Regulation, Wonkwang University School of Medicine, Iksan, Korea
| | - Raekil Park
- Department of Biomedical Science & Engineering, Gwangju Institute of Science & Technology, Gwangju, Korea
| |
Collapse
|
9
|
Menzies SA, Volkmar N, van den Boomen DJH, Timms RT, Dickson AS, Nathan JA, Lehner PJ. The sterol-responsive RNF145 E3 ubiquitin ligase mediates the degradation of HMG-CoA reductase together with gp78 and Hrd1. eLife 2018; 7:e40009. [PMID: 30543180 PMCID: PMC6292692 DOI: 10.7554/elife.40009] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/19/2018] [Indexed: 02/02/2023] Open
Abstract
Mammalian HMG-CoA reductase (HMGCR), the rate-limiting enzyme of the cholesterol biosynthetic pathway and the therapeutic target of statins, is post-transcriptionally regulated by sterol-accelerated degradation. Under cholesterol-replete conditions, HMGCR is ubiquitinated and degraded, but the identity of the E3 ubiquitin ligase(s) responsible for mammalian HMGCR turnover remains controversial. Using systematic, unbiased CRISPR/Cas9 genome-wide screens with a sterol-sensitive endogenous HMGCR reporter, we comprehensively map the E3 ligase landscape required for sterol-accelerated HMGCR degradation. We find that RNF145 and gp78 independently co-ordinate HMGCR ubiquitination and degradation. RNF145, a sterol-responsive ER-resident E3 ligase, is unstable but accumulates following sterol depletion. Sterol addition triggers RNF145 recruitment to HMGCR via Insigs, promoting HMGCR ubiquitination and proteasome-mediated degradation. In the absence of both RNF145 and gp78, Hrd1, a third UBE2G2-dependent E3 ligase, partially regulates HMGCR activity. Our findings reveal a critical role for the sterol-responsive RNF145 in HMGCR regulation and elucidate the complexity of sterol-accelerated HMGCR degradation. Editorial note This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).
Collapse
Affiliation(s)
- Sam A Menzies
- Department of MedicineCambridge Institute for Medical ResearchCambridgeUnited Kingdom
| | - Norbert Volkmar
- Department of MedicineCambridge Institute for Medical ResearchCambridgeUnited Kingdom
| | | | - Richard T Timms
- Department of MedicineCambridge Institute for Medical ResearchCambridgeUnited Kingdom
| | - Anna S Dickson
- Department of MedicineCambridge Institute for Medical ResearchCambridgeUnited Kingdom
| | - James A Nathan
- Department of MedicineCambridge Institute for Medical ResearchCambridgeUnited Kingdom
| | - Paul J Lehner
- Department of MedicineCambridge Institute for Medical ResearchCambridgeUnited Kingdom
| |
Collapse
|
10
|
Shen L, Tenzer S, Hess M, Distler U, Tubbe I, Montermann E, Schimmer S, Dittmer U, Grabbe S, Bros M. Friend virus limits adaptive cellular immune responses by imprinting a maturation-resistant and T helper type 2-biased immunophenotype in dendritic cells. PLoS One 2018; 13:e0192541. [PMID: 29425215 PMCID: PMC5806892 DOI: 10.1371/journal.pone.0192541] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 01/25/2018] [Indexed: 12/28/2022] Open
Abstract
The murine Friend virus (FV) retrovirus model has been widely used to study anti-viral immune responses, and virus-induced cancer. Here we analyzed FV immune evasion mechanisms on the level of dendritic cells (DC) essential for the induction of primary adaptive immune responses. Comparative quantitative proteome analysis of FV-infected DC (FV-DC) of different genotypes (BALB/c, C57BL/6) and non-infected DC revealed numerous genotype-independently regulated proteins rergulating metabolic activity, cytoskeletal rearrangements, and antigen processing/presentation. These alterations may promote virion production in FV-DC. Stimulation of FV-DC with LPS resulted in strongly enhanced IL-10 production which was partially responsible for their attenuated T cell (CD4+, CD8+) stimulatory capacity. Stimulated FV-DC induced less IFN-γ production in T cells required for cellular anti-viral responses, but more T helper cell type 2 (Th2)-associated cytokines (IL-4, IL-5, IL-13). We conclude that FV reprograms DC to promote viral spreading and immune deviation by imprinting a largely maturation-resistant, Th2-biased immunophenotype.
Collapse
Affiliation(s)
- Limei Shen
- Department of Dermatology, University Medical Center, Mainz, Germany
| | - Stefan Tenzer
- Institute of Immunology, University Medical Center, Mainz, Germany
| | - Moritz Hess
- Institute for Medical Biometry, Epidemiology and Informatics, University Medical Center, Mainz, Germany
| | - Ute Distler
- Institute of Immunology, University Medical Center, Mainz, Germany
| | - Ingrid Tubbe
- Department of Dermatology, University Medical Center, Mainz, Germany
| | - Evelyn Montermann
- Department of Dermatology, University Medical Center, Mainz, Germany
| | - Simone Schimmer
- Institute for Virology of the University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulf Dittmer
- Institute for Virology of the University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center, Mainz, Germany
- * E-mail:
| | - Matthias Bros
- Department of Dermatology, University Medical Center, Mainz, Germany
| |
Collapse
|
11
|
Mixture effects of azole fungicides on the adrenal gland in a broad dose range. Toxicology 2017; 385:28-37. [DOI: 10.1016/j.tox.2017.04.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/07/2017] [Accepted: 04/24/2017] [Indexed: 02/04/2023]
|
12
|
Chang CW, Abhinav K, Di Cara F, Panagakou I, Vass S, Heck MMS. A role for the metalloprotease invadolysin in insulin signaling and adipogenesis. Biol Chem 2016; 398:373-393. [PMID: 27622830 DOI: 10.1515/hsz-2016-0226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/04/2016] [Indexed: 01/24/2023]
Abstract
Invadolysin is a novel metalloprotease conserved amongst metazoans that is essential for life in Drosophila. We previously showed that invadolysin was essential for the cell cycle and cell migration, linking to metabolism through a role in lipid storage and interaction with mitochondrial proteins. In this study we demonstrate that invadolysin mutants exhibit increased autophagy and decreased glycogen storage - suggestive of a role for invadolysin in insulin signaling in Drosophila. Consistent with this, effectors of insulin signaling were decreased in invadolysin mutants. In addition, we discovered that invadolysin was deposited on newly synthesized lipid droplets in a PKC-dependent manner. We examined two in vitro models of adipogenesis for the expression and localization of invadolysin. The level of invadolysin increased during both murine 3T3-L1 and human Simpson-Golabi-Behmel syndrome (SGBS), adipogenesis. Invadolysin displayed a dynamic localization to lipid droplets over the course of adipogenesis, which may be due to the differential expression of distinct invadolysin variants. Pharmacological inhibition of adipogenesis abrogated the increase in invadolysin. In summary, our results on in vivo and in vitro systems highlight an important role for invadolysin in insulin signaling and adipogenesis.
Collapse
|
13
|
Thakur V, Asad M, Jain S, Hossain ME, Gupta A, Kaur I, Rathore S, Ali S, Khan NJ, Mohmmed A. Eps15 homology domain containing protein of Plasmodium falciparum (PfEHD) associates with endocytosis and vesicular trafficking towards neutral lipid storage site. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2856-69. [PMID: 26284889 DOI: 10.1016/j.bbamcr.2015.08.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 07/19/2015] [Accepted: 08/07/2015] [Indexed: 01/08/2023]
Abstract
The human malaria parasite, Plasmodium falciparum, takes up numerous host cytosolic components and exogenous nutrients through endocytosis during the intra-erythrocytic stages. Eps15 homology domain-containing proteins (EHDs) are conserved NTPases, which are implicated in membrane remodeling and regulation of specific endocytic transport steps in eukaryotic cells. In the present study, we have characterized the dynamin-like C-terminal Eps15 homology domain containing protein of P. falciparum (PfEHD). Using a GFP-targeting approach, we studied localization and trafficking of PfEHD in the parasite. The PfEHD-GFP fusion protein was found to be a membrane bound protein that associates with vesicular network in the parasite. Time-lapse microscopy studies showed that these vesicles originate at parasite plasma membrane, migrate through the parasite cytosol and culminate into a large multi-vesicular like structure near the food-vacuole. Co-staining of food vacuole membrane showed that the multi-vesicular structure is juxtaposed but outside the food vacuole. Labeling of parasites with neutral lipid specific dye, Nile Red, showed that this large structure is neutral lipid storage site in the parasites. Proteomic analysis identified endocytosis modulators as PfEHD associated proteins in the parasites. Treatment of parasites with endocytosis inhibitors obstructed the development of PfEHD-labeled vesicles and blocked their targeting to the lipid storage site. Overall, our data suggests that the PfEHD is involved in endocytosis and plays a role in the generation of endocytic vesicles at the parasite plasma membrane, that are subsequently targeted to the neutral lipid generation/storage site localized near the food vacuole.
Collapse
Affiliation(s)
- Vandana Thakur
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India
| | - Mohd Asad
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India; Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110 025, India
| | - Shaifali Jain
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India
| | - Mohammad E Hossain
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India
| | - Akanksha Gupta
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India
| | - Inderjeet Kaur
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India
| | - Sumit Rathore
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi 110 029, India
| | - Shakir Ali
- Department of Biochemistry, Faculty of Science, Jamia Hamdard, New Delhi 110062, India
| | - Nida J Khan
- Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110 025, India
| | - Asif Mohmmed
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110 067, India.
| |
Collapse
|
14
|
Proteomic analysis of murine testes lipid droplets. Sci Rep 2015; 5:12070. [PMID: 26159641 PMCID: PMC4498221 DOI: 10.1038/srep12070] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 06/10/2015] [Indexed: 01/12/2023] Open
Abstract
Testicular Leydig cells contain abundant cytoplasmic lipid droplets (LDs) as a cholesteryl-ester store for releasing cholesterols as the precursor substrate for testosterone biosynthesis. Here, we identified the protein composition of testicular LDs purified from adult mice by using mass spectrometry and immunodetection. Among 337 proteins identified, 144 were previously detected in LD proteomes; 44 were confirmed by microscopy. Testicular LDs contained multiple Rab GTPases, chaperones, and proteins involved in glucuronidation, ubiquination and transport, many known to modulate LD formation and LD-related cellular functions. In particular, testicular LDs contained many members of both the perilipin family and classical lipase/esterase superfamily assembled predominately in adipocyte LDs. Thus, testicular LDs might be regulated similar to adipocyte LDs. Remarkably, testicular LDs contained a large number of classical enzymes for biosynthesis and metabolism of cholesterol and hormonal steroids, so steroidogenic reactions might occur on testicular LDs or the steroidogenic enzymes and products could be transferred through testicular LDs. These characteristics differ from the LDs in most other types of cells, so testicular LDs could be an active organelle functionally involved in steroidogenesis.
Collapse
|
15
|
Liu Y, Chen H, Sun Z, Chen X. Molecular mechanisms of ethanol-associated oro-esophageal squamous cell carcinoma. Cancer Lett 2015; 361:164-173. [PMID: 25766659 PMCID: PMC4765374 DOI: 10.1016/j.canlet.2015.03.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/04/2015] [Accepted: 03/04/2015] [Indexed: 02/06/2023]
Abstract
Alcohol drinking is a major etiological factor of oro-esophageal squamous cell carcinoma (OESCC). Both local and systemic effects of ethanol may promote carcinogenesis, especially among chronic alcoholics. However, molecular mechanisms of ethanol-associated OESCC are still not well understood. In this review, we summarize current understandings and propose three mechanisms of ethanol-associated OESCC: (1) Disturbance of systemic metabolism of nutrients: during ethanol metabolism in the liver, systemic metabolism of retinoids, zinc, iron and methyl groups is altered. These nutrients are known to be associated with the development of OESCC. (2) Disturbance of redox metabolism in squamous epithelial cells: when ethanol is metabolized in oro-esophageal squamous epithelial cells, reactive oxygen species are generated and produce oxidative damage. Meanwhile, ethanol may also disturb fatty-acid metabolism in these cells. (3) Disturbance of signaling pathways in squamous epithelial cells: due to its physico-chemical properties, ethanol changes cell membrane fluidity and shape, and may thus impact multiple signaling pathways. Advanced molecular techniques in genomics, epigenomics, metabolomics and microbiomics will help us elucidate how ethanol promotes OESCC.
Collapse
Affiliation(s)
- Yao Liu
- Department of Oral Medicine, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China; Cancer Research Program, JLC-BBRI, North Carolina Central University, Durham, NC 27707, USA
| | - Hao Chen
- Cancer Research Program, JLC-BBRI, North Carolina Central University, Durham, NC 27707, USA
| | - Zheng Sun
- Department of Oral Medicine, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China.
| | - Xiaoxin Chen
- Cancer Research Program, JLC-BBRI, North Carolina Central University, Durham, NC 27707, USA.
| |
Collapse
|
16
|
Posey AD, Swanson KE, Alvarez MG, Krishnan S, Earley JU, Band H, Pytel P, McNally EM, Demonbreun AR. EHD1 mediates vesicle trafficking required for normal muscle growth and transverse tubule development. Dev Biol 2014; 387:179-90. [PMID: 24440153 DOI: 10.1016/j.ydbio.2014.01.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 01/06/2014] [Accepted: 01/07/2014] [Indexed: 01/03/2023]
Abstract
EHD proteins have been implicated in intracellular trafficking, especially endocytic recycling, where they mediate receptor and lipid recycling back to the plasma membrane. Additionally, EHDs help regulate cytoskeletal reorganization and induce tubule formation. It was previously shown that EHD proteins bind directly to the C2 domains in myoferlin, a protein that regulates myoblast fusion. Loss of myoferlin impairs normal myoblast fusion leading to smaller muscles in vivo but the intracellular pathways perturbed by loss of myoferlin function are not well known. We now characterized muscle development in EHD1-null mice. EHD1-null myoblasts display defective receptor recycling and mislocalization of key muscle proteins, including caveolin-3 and Fer1L5, a related ferlin protein homologous to myoferlin. Additionally, EHD1-null myoblast fusion is reduced. We found that loss of EHD1 leads to smaller muscles and myofibers in vivo. In wildtype skeletal muscle EHD1 localizes to the transverse tubule (T-tubule), and loss of EHD1 results in overgrowth of T-tubules with excess vesicle accumulation in skeletal muscle. We provide evidence that tubule formation in myoblasts relies on a functional EHD1 ATPase domain. Moreover, we extended our studies to show EHD1 regulates BIN1 induced tubule formation. These data, taken together and with the known interaction between EHD and ferlin proteins, suggests that the EHD proteins coordinate growth and development likely through mediating vesicle recycling and the ability to reorganize the cytoskeleton.
Collapse
Affiliation(s)
- Avery D Posey
- Committee on Genetics, Genomics and Systems Biology, The University of Chicago, Chicago, IL 60637, USA
| | - Kaitlin E Swanson
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Manuel G Alvarez
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Swathi Krishnan
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Judy U Earley
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Hamid Band
- Eppley Institute for Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Peter Pytel
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Elizabeth M McNally
- Committee on Genetics, Genomics and Systems Biology, The University of Chicago, Chicago, IL 60637, USA; Department of Medicine, The University of Chicago, Chicago, IL 60637, USA; Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
17
|
Simone LC, Naslavsky N, Caplan S. Scratching the surface: actin' and other roles for the C-terminal Eps15 homology domain protein, EHD2. Histol Histopathol 2013; 29:285-92. [PMID: 24347515 DOI: 10.14670/hh-29.285] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The C-terminal Eps15 homology domain-containing (EHD) proteins participate in multiple aspects of endocytic membrane trafficking. Of the four mammalian EHD proteins, EHD2 appears to be the most disparate, both in terms of sequence homology, and in subcellular localization/function. Since its initial description as a plasma membrane-associated protein, the precise function of EHD2 has remained enigmatic. Various reports have suggested roles for EHD2 at the plasma membrane, within the endocytic transport system, and even in the nucleus. For example, EHD2 facilitates membrane fusion/repair in muscle cells. Recently the focus has shifted to the role of EHD2 in regulating caveolae. Indeed, EHD2 is highly expressed in tissues rich in caveolae, including fat, muscle and blood vessels. This review highlights cumulative evidence linking EHD2 to actin-rich structures at the plasma membrane, where the plasma membrane-associated phospholipid phosphatidylinositol 4,5-bisphosphate controls EHD2 recruitment. Herein we examine the key pathways where EHD2 might function, and address its potential involvement in these processes.
Collapse
Affiliation(s)
- Laura C Simone
- Department of Biochemistry and Molecular Biology and the Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Naava Naslavsky
- Department of Biochemistry and Molecular Biology and the Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Steve Caplan
- Department of Biochemistry and Molecular Biology and the Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
18
|
Lu H, Meng Q, Wen Y, Hu J, Zhao Y, Cai L. Increased EHD1 in non-small cell lung cancer predicts poor survival. Thorac Cancer 2013; 4:422-432. [PMID: 28920217 DOI: 10.1111/1759-7714.12043] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 03/24/2013] [Indexed: 11/28/2022] Open
Affiliation(s)
- Hailing Lu
- The Forth Department of Medical Oncology; The Tumor Hospital of Harbin Medical University; Harbin China
| | - Qingwei Meng
- The Forth Department of Medical Oncology; The Tumor Hospital of Harbin Medical University; Harbin China
| | - Yuan Wen
- The Forth Department of Medical Oncology; The Tumor Hospital of Harbin Medical University; Harbin China
| | - Jing Hu
- The Forth Department of Medical Oncology; The Tumor Hospital of Harbin Medical University; Harbin China
| | - Yanbin Zhao
- The Forth Department of Medical Oncology; The Tumor Hospital of Harbin Medical University; Harbin China
| | - Li Cai
- The Forth Department of Medical Oncology; The Tumor Hospital of Harbin Medical University; Harbin China
| |
Collapse
|
19
|
Deracinois B, Duban-Deweer S, Pottiez G, Cecchelli R, Karamanos Y, Flahaut C. TNAP and EHD1 are over-expressed in bovine brain capillary endothelial cells after the re-induction of blood-brain barrier properties. PLoS One 2012; 7:e48428. [PMID: 23119012 PMCID: PMC3485243 DOI: 10.1371/journal.pone.0048428] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 09/25/2012] [Indexed: 12/31/2022] Open
Abstract
Although the physiological properties of the blood-brain barrier (BBB) are relatively well known, the phenotype of the component brain capillary endothelial cells (BCECs) has yet to be described in detail. Likewise, the molecular mechanisms that govern the establishment and maintenance of the BBB are largely unknown. Proteomics can be used to assess quantitative changes in protein levels and identify proteins involved in the molecular pathways responsible for cellular differentiation. Using the well-established in vitro BBB model developed in our laboratory, we performed a differential nano-LC MALDI-TOF/TOF-MS study of Triton X-100-soluble protein species from bovine BCECs displaying either limited BBB functions or BBB functions re-induced by glial cells. Due to the heterogeneity of the crude extract, we increased identification yields by applying a repeatable, reproducible fractionation process based on the proteins' relative hydrophobicity. We present proteomic and biochemical evidence to show that tissue non-specific alkaline phosphatase (TNAP) and Eps15 homology domain-containing protein 1(EDH1) are over-expressed by bovine BCECs after the re-induction of BBB properties. We discuss the impact of these findings on current knowledge of endothelial and BBB permeability.
Collapse
Affiliation(s)
- Barbara Deracinois
- Université Lille Nord de France, Lille, France
- Université d’Artois, LBHE, Lens, France
- IMPRT-IFR114, Lille, France
| | - Sophie Duban-Deweer
- Université Lille Nord de France, Lille, France
- Université d’Artois, LBHE, Lens, France
- IMPRT-IFR114, Lille, France
| | - Gwënaël Pottiez
- Université Lille Nord de France, Lille, France
- Université d’Artois, LBHE, Lens, France
- IMPRT-IFR114, Lille, France
| | - Roméo Cecchelli
- Université Lille Nord de France, Lille, France
- Université d’Artois, LBHE, Lens, France
- IMPRT-IFR114, Lille, France
| | - Yannis Karamanos
- Université Lille Nord de France, Lille, France
- Université d’Artois, LBHE, Lens, France
- IMPRT-IFR114, Lille, France
| | - Christophe Flahaut
- Université Lille Nord de France, Lille, France
- Université d’Artois, LBHE, Lens, France
- IMPRT-IFR114, Lille, France
- * E-mail:
| |
Collapse
|
20
|
Moir RD, Gross DA, Silver DL, Willis IM. SCS3 and YFT2 link transcription of phospholipid biosynthetic genes to ER stress and the UPR. PLoS Genet 2012; 8:e1002890. [PMID: 22927826 PMCID: PMC3426550 DOI: 10.1371/journal.pgen.1002890] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 06/19/2012] [Indexed: 11/21/2022] Open
Abstract
The ability to store nutrients in lipid droplets (LDs) is an ancient function that provides the primary source of metabolic energy during periods of nutrient insufficiency and between meals. The Fat storage-Inducing Transmembrane (FIT) proteins are conserved ER–resident proteins that facilitate fat storage by partitioning energy-rich triglycerides into LDs. FIT2, the ancient ortholog of the FIT gene family first identified in mammals has two homologs in Saccharomyces cerevisiae (SCS3 and YFT2) and other fungi of the Saccharomycotina lineage. Despite the coevolution of these genes for more than 170 million years and their divergence from higher eukaryotes, SCS3, YFT2, and the human FIT2 gene retain some common functions: expression of the yeast genes in a human embryonic kidney cell line promotes LD formation, and expression of human FIT2 in yeast rescues the inositol auxotrophy and chemical and genetic phenotypes of strains lacking SCS3. To better understand the function of SCS3 and YFT2, we investigated the chemical sensitivities of strains deleted for either or both genes and identified synthetic genetic interactions against the viable yeast gene-deletion collection. We show that SCS3 and YFT2 have shared and unique functions that connect major biosynthetic processes critical for cell growth. These include lipid metabolism, vesicular trafficking, transcription of phospholipid biosynthetic genes, and protein synthesis. The genetic data indicate that optimal strain fitness requires a balance between phospholipid synthesis and protein synthesis and that deletion of SCS3 and YFT2 impacts a regulatory mechanism that coordinates these processes. Part of this mechanism involves a role for SCS3 in communicating changes in the ER (e.g. due to low inositol) to Opi1-regulated transcription of phospholipid biosynthetic genes. We conclude that SCS3 and YFT2 are required for normal ER membrane biosynthesis in response to perturbations in lipid metabolism and ER stress. The ability to form lipid droplets is a conserved property of eukaryotic cells that allows the storage of excess metabolic energy in a form that can be readily accessed. In adipose tissue, the storage of excess calories in lipid droplets normally protects other tissues from lipotoxicity and insulin resistance, but this protection is lost with chronic over-nutrition. The FAT storage-inducing transmembrane (FIT) proteins were recently identified as a conserved family of proteins that reside in the lipid bilayer of the endoplasmic reticulum and are implicated in lipid droplet formation. In this work we show that specific functions of the FIT proteins are conserved between yeast and humans and that SCS3 and YFT2, the yeast homologs of mammalian FIT2, are part of a large genetic interaction network connecting lipid metabolism, vesicle trafficking, transcription, and protein synthesis. From these interactions we determined that yeast strains lacking SCS3 and YFT2 are defective in their response to chronic ER stress and cannot induce the unfolded protein response pathway or transcription of phospholipid biosynthetic genes in low inositol. Our findings suggest that the mammalian FIT genes may play an important role in ER stress pathways, which are linked to obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Robyn D. Moir
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - David A. Gross
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke–NUS Graduate Medical School Singapore, Singapore, Singapore
| | - David L. Silver
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Signature Research Program in Cardiovascular and Metabolic Disorders, Duke–NUS Graduate Medical School Singapore, Singapore, Singapore
- * E-mail: (IMW); (DLS)
| | - Ian M. Willis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail: (IMW); (DLS)
| |
Collapse
|
21
|
Murphy DJ. The dynamic roles of intracellular lipid droplets: from archaea to mammals. PROTOPLASMA 2012; 249:541-85. [PMID: 22002710 DOI: 10.1007/s00709-011-0329-7] [Citation(s) in RCA: 271] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 09/28/2011] [Indexed: 05/02/2023]
Abstract
During the past decade, there has been a paradigm shift in our understanding of the roles of intracellular lipid droplets (LDs). New genetic, biochemical and imaging technologies have underpinned these advances, which are revealing much new information about these dynamic organelles. This review takes a comparative approach by examining recent work on LDs across the whole range of biological organisms from archaea and bacteria, through yeast and Drosophila to mammals, including humans. LDs probably evolved originally in microorganisms as temporary stores of excess dietary lipid that was surplus to the immediate requirements of membrane formation/turnover. LDs then acquired roles as long-term carbon stores that enabled organisms to survive episodic lack of nutrients. In multicellular organisms, LDs went on to acquire numerous additional roles including cell- and organism-level lipid homeostasis, protein sequestration, membrane trafficking and signalling. Many pathogens of plants and animals subvert their host LD metabolism as part of their infection process. Finally, malfunctions in LDs and associated proteins are implicated in several degenerative diseases of modern humans, among the most serious of which is the increasingly prevalent constellation of pathologies, such as obesity and insulin resistance, which is associated with metabolic syndrome.
Collapse
Affiliation(s)
- Denis J Murphy
- Division of Biological Sciences, University of Glamorgan, Cardiff, CF37 4AT, UK.
| |
Collapse
|
22
|
Zhang J, Reiling C, Reinecke JB, Prislan I, Marky LA, Sorgen PL, Naslavsky N, Caplan S. Rabankyrin-5 interacts with EHD1 and Vps26 to regulate endocytic trafficking and retromer function. Traffic 2012; 13:745-57. [PMID: 22284051 DOI: 10.1111/j.1600-0854.2012.01334.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 01/24/2012] [Accepted: 01/27/2012] [Indexed: 12/13/2022]
Abstract
Rabankyrin-5 (Rank-5) has been implicated as an effector of the small GTPase Rab5 and plays an important role in macropinocytosis. We have now identified Rank-5 as an interaction partner for the recycling regulatory protein, Eps15 homology domain 1 (EHD1). We have demonstrated this interaction by glutathione S-transferase-pulldown, yeast two-hybrid assay, isothermal calorimetry and co-immunoprecipitation, and found that the binding occurs between the EH domain of EHD1 and the NPFED motif of Rank-5. Similar to EHD1, we found that Rank-5 colocalizes and interacts with components of the retromer complex such as vacuolar protein sorting 26 (Vps26), suggesting a role for Rank-5 in retromer-based transport. Indeed, depletion of Rank-5 causes mislocalization of Vps26 and affects both the retrieval of mannose 6-phosphate receptor transport to the Golgi from endosomes and biosynthetic transport. Moreover, Rank-5 is required for normal retromer distribution, as overexpression of a wild-type Rank-5-small interfering RNA-resistant construct rescues retromer mislocalization. Finally, we show that depletion of either Rank-5 or EHD1 impairs secretion of vesicular stomatitis virus glycoprotein. Overall, our data identify a new interaction between Rank-5 and EHD1, and novel endocytic regulatory roles that include retromer-based transport and secretion.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Biochemistry and Molecular Biology and Eppley Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Peränen J. Rab8 GTPase as a regulator of cell shape. Cytoskeleton (Hoboken) 2011; 68:527-39. [PMID: 21850707 DOI: 10.1002/cm.20529] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 08/08/2011] [Accepted: 08/09/2011] [Indexed: 12/14/2022]
Abstract
Endogenous Rab8 is found in dynamic cell structures like filopodia, lamellipodia, protrusions, ruffles, and primary cilia. Activation of Rab8 is linked to the formation of these actin containing structures, whereas inhibition of Rab8 affects negatively their appearance. The activity of Rab8 is controlled by specific guanine nucleotide exchange factors and GTPase activating proteins. Rab8 regulates a membrane recycling pathway that is linked to Arf6, EHD1, Myo5, and Rab11. A hypothesis is presented on the role of Rab8 in the formation of new cell surface domains. The review focuses on the function of Rab8 in cell migration, epithelial polarization, neuron differentiation, and ciliogenesis.
Collapse
Affiliation(s)
- Johan Peränen
- Institute of Biotechnology, University of Helsinki, Finland.
| |
Collapse
|
24
|
Joo JI, Oh TS, Kim DH, Choi DK, Wang X, Choi JW, Yun JW. Differential expression of adipose tissue proteins between obesity-susceptible and -resistant rats fed a high-fat diet. Proteomics 2011; 11:1429-48. [PMID: 21365757 DOI: 10.1002/pmic.201000515] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 11/17/2010] [Accepted: 01/12/2011] [Indexed: 01/10/2023]
Abstract
One of the major questions in the field of obesity is why some humans become obese (obesity prone, OP) and others resist the development of obesity (obesity resistant, OR) when exposed to a high-calorie diet, which has not been completely studied. Therefore, in the present study, in order to gain insight into the molecular mechanisms underlying this propensity, we have performed a comparative analysis of protein expression profiles in white adipose tissue (WAT) and brown adipose tissue (BAT) of rats fed a high-fat diet by 2-DE and MALDI-TOF-MS. Protein mapping of homogenates revealed significant alterations to a number of proteins; 60 and 70 proteins were differentially regulated in BAT and WAT, respectively. For careful interpretation of proteomic results, we categorized the identified proteins into two groups by analysis of both average spot density of pooled six rat adipose tissues and individual spot density of each adipose tissue of six rats as a function of body weight. One of the most striking findings of this study was that significant changes of Ehd1 and laminin receptor in BAT as well as antiquitin, DJ-1 protein, and paraoxonase 2 in WAT were found for the first time in obese rats. In addition, we confirmed the increased expression of some thermogenic enzymes and decreased lipogenic enzymes in adipose tissues of OR rats by immunoblot analysis. To our knowledge, this is the first proteomic study of profiling of protein modulation in OP and OR rats, thereby providing the first global evidence for different propensities to obesity between OP and OR rats.
Collapse
Affiliation(s)
- Jeong In Joo
- Department of Biotechnology, Daegu University, Kyungsan, Kyungbuk, Korea
| | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Myoblast fusion contributes to muscle growth in development and during regeneration of mature muscle. Myoblasts fuse to each other as well as to multinucleate myotubes to enlarge the myofiber. The molecular mechanisms of myoblast fusion are incompletely understood. Adhesion, apposition, and membrane fusion are accompanied by cytoskeletal rearrangements. The ferlin family of proteins is implicated in human muscle disease and has been implicated in fusion events in muscle, including myoblast fusion, vesicle trafficking and membrane repair. Dysferlin was the first mammalian ferlin identified and it is now known that there are six different ferlins. Loss-of-function mutations in the dysferlin gene lead to limb girdle muscular dystrophy and the milder disorder Miyoshi Myopathy. Dysferlin is a membrane-associated protein that has been implicated in resealing disruptions in the muscle plasma membrane. Newer data supports a broader role for dysferlin in intracellular vesicular movement, a process also important for resealing. Myoferlin is highly expressed in myoblasts that undergoing fusion, and the absence of myoferlin leads to impaired myoblast fusion. Myoferlin also regulates intracellular trafficking events, including endocytic recycling, a process where internalized vesicles are returned to the plasma membrane. The trafficking role of ferlin proteins is reviewed herein with a specific focus as to how this machinery alters myogenesis and muscle growth.
Collapse
Affiliation(s)
- Avery D Posey
- Genomics and Systems Biology, Committee on Genetics, The University of Chicago, Chicago, Illinois, USA
| | | | | |
Collapse
|
26
|
Cai B, Katafiasz D, Horejsi V, Naslavsky N. Pre-sorting endosomal transport of the GPI-anchored protein, CD59, is regulated by EHD1. Traffic 2010; 12:102-20. [PMID: 20961375 DOI: 10.1111/j.1600-0854.2010.01135.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
EHD1 regulates the trafficking of multiple receptors from the endocytic recycling compartment (ERC) to the plasma membrane. However, the potential role of EHD1 in regulating the family of glycosylphosphatidylinositol-anchored proteins (GPI-APs) has not been determined. Here we demonstrate a novel role for EHD1 in regulating the trafficking of CD59, an endogenous GPI-AP, at early stages of trafficking through the endocytic pathway. EHD1 displays significant colocalization with newly internalized CD59. Upon EHD1 depletion, there is a rapid Rab5-independent coalescence of CD59 in the ERC region. However, expression of an active Arf6 mutant (Q67L), which traps internalized pre-sorting endosomal cargo in phosphatidylinositol(4,5)-bisphosphate enriched vacuoles, prevents this coalescence. It is of interest that sustained PKC activation leads to a similar coalescence of CD59 at the ERC, and treatment of EHD1-depleted cells with a PKC inhibitor (Go6976) blocked this rapid relocation of CD59. However, unlike sustained PKC activation, EHD1 depletion does not induce the translocation of PKCα to ERC. The results presented herein provide evidence that EHD1 is involved in the control of CD59 transport from pre-sorting endosomes to the ERC in a PKC-dependent manner. However, the mechanisms of EHD1-induced coalescence of CD59 at the ERC differ from those induced by sustained PKC activation.
Collapse
Affiliation(s)
- Bishuang Cai
- Department of Biochemistry and Molecular Biology and Eppley Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | | | | | | |
Collapse
|
27
|
EHD proteins: key conductors of endocytic transport. Trends Cell Biol 2010; 21:122-31. [PMID: 21067929 DOI: 10.1016/j.tcb.2010.10.003] [Citation(s) in RCA: 196] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Revised: 10/07/2010] [Accepted: 10/07/2010] [Indexed: 12/12/2022]
Abstract
Regulation of endocytic transport is controlled by an elaborate network of proteins. Rab GTP-binding proteins and their effectors have well-defined roles in mediating specific endocytic transport steps, but until recently less was known about the four mammalian dynamin-like C-terminal Eps15 homology domain (EHD) proteins that also regulate endocytic events. In recent years, however, great strides have been made in understanding the structure and function of these unique proteins. Indeed, a growing body of literature addresses EHD protein structure, interactions with binding partners, functions in mammalian cells, and the generation of various new model systems. Accordingly, this is now an opportune time to pause and review the function and mechanisms of action of EHD proteins, and to highlight some of the challenges and future directions for the field.
Collapse
|
28
|
Rahajeng J, Giridharan SSP, Cai B, Naslavsky N, Caplan S. Important relationships between Rab and MICAL proteins in endocytic trafficking. World J Biol Chem 2010; 1:254-64. [PMID: 21537482 PMCID: PMC3083971 DOI: 10.4331/wjbc.v1.i8.254] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Revised: 07/28/2010] [Accepted: 08/04/2010] [Indexed: 02/05/2023] Open
Abstract
The internalization of essential nutrients, lipids and receptors is a crucial process for all eukaryotic cells. Accordingly, endocytosis is highly conserved across cell types and species. Once internalized, small cargo-containing vesicles fuse with early endosomes (also known as sorting endosomes), where they undergo segregation to distinct membrane regions and are sorted and transported on through the endocytic pathway. Although the mechanisms that regulate this sorting are still poorly understood, some receptors are directed to late endosomes and lysosomes for degradation, whereas other receptors are recycled back to the plasma membrane; either directly or through recycling endosomes. The Rab family of small GTP-binding proteins plays crucial roles in regulating these trafficking pathways. Rabs cycle from inactive GDP-bound cytoplasmic proteins to active GTP-bound membrane-associated proteins, as a consequence of the activity of multiple specific GTPase-activating proteins (GAPs) and GTP exchange factors (GEFs). Once bound to GTP, Rabs interact with a multitude of effector proteins that carry out Rab-specific functions. Recent studies have shown that some of these effectors are also interaction partners for the C-terminal Eps15 homology (EHD) proteins, which are also intimately involved in endocytic regulation. A particularly interesting example of common Rab-EHD interaction partners is the MICAL-like protein, MICAL-L1. MICAL-L1 and its homolog, MICAL-L2, belong to the larger MICAL family of proteins, and both have been directly implicated in regulating endocytic recycling of cell surface receptors and junctional proteins, as well as controlling cytoskeletal rearrangement and neurite outgrowth. In this review, we summarize the functional roles of MICAL and Rab proteins, and focus on the significance of their interactions and the implications for endocytic transport.
Collapse
Affiliation(s)
- Juliati Rahajeng
- Juliati Rahajeng, Sai Srinivas Panapakkam Giridharan, Bishuang Cai, Naava Naslavsky, Steve Caplan, Department of Biochemistry and Molecular Biology, and Eppley Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870, United States
| | | | | | | | | |
Collapse
|
29
|
George M, Rainey MA, Naramura M, Ying G, Harms DW, Vitaterna MH, Doglio L, Crawford SE, Hess RA, Band V, Band H. Ehd4 is required to attain normal prepubertal testis size but dispensable for fertility in male mice. Genesis 2010; 48:328-42. [PMID: 20213691 DOI: 10.1002/dvg.20620] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The four highly homologous members of the C-terminal EH domain-containing (EHD) protein family (EHD1-4) regulate endocytic recycling. To delineate the role of EHD4 in normal physiology and development, mice with a conditional knockout of the Ehd4 gene were generated. PCR of genomic DNA and Western blotting of organ lysates from Ehd4(-/-) mice confirmed EHD4 deletion. Ehd4(-/-) mice were viable and born at expected Mendelian ratios; however, males showed a 50% reduction in testis weight, obvious from postnatal day 31. An early (Day 10) increase in germ cell proliferation and apoptosis and a later increase in apoptosis (Day 31) were seen in the Ehd4(-/-) testis. Other defects included a progressive reduction in seminiferous tubule diameter, dysregulation of seminiferous epithelium, and head abnormalities in elongated spermatids. As a consequence, lower sperm counts and reduced fertility were observed in Ehd4(-/-) males. Interestingly, EHD protein expression was seen to be temporally regulated in the testis and EHD4 levels peaked between days 10 and 15. In the adult testis, EHD4 was highly expressed in primary spermatocytes and EHD4 deletion altered the levels of other EHD proteins in an age-dependent manner. We conclude that high levels of EHD1 in the adult Ehd4(-/-) testis functionally compensate for lack of EHD4 and prevents the development of severe fertility defects. Our results suggest a role for EHD4 in the proper development of postmitotic and postmeiotic germ cells and implicate EHD protein-mediated endocytic recycling as an important process in germ cell development and testis function.
Collapse
Affiliation(s)
- Manju George
- Eppley Institute for Research in Cancer and Allied Diseases, UNMC-Eppley Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Gudmundsson H, Hund TJ, Wright PJ, Kline CF, Snyder JS, Qian L, Koval OM, Cunha SR, George M, Rainey MA, Kashef FE, Dun W, Boyden PA, Anderson ME, Band H, Mohler PJ. EH domain proteins regulate cardiac membrane protein targeting. Circ Res 2010; 107:84-95. [PMID: 20489164 DOI: 10.1161/circresaha.110.216713] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
RATIONALE Cardiac membrane excitability is tightly regulated by an integrated network of membrane-associated ion channels, transporters, receptors, and signaling molecules. Membrane protein dynamics in health and disease are maintained by a complex ensemble of intracellular targeting, scaffolding, recycling, and degradation pathways. Surprisingly, despite decades of research linking dysfunction in membrane protein trafficking with human cardiovascular disease, essentially nothing is known regarding the molecular identity or function of these intracellular targeting pathways in excitable cardiomyocytes. OBJECTIVE We sought to discover novel pathways for membrane protein targeting in primary cardiomyocytes. METHODS AND RESULTS We report the initial characterization of a large family of membrane trafficking proteins in human heart. We used a tissue-wide screen for novel ankyrin-associated trafficking proteins and identified 4 members of a unique Eps15 homology (EH) domain-containing protein family (EHD1, EHD2, EHD3, EHD4) that serve critical roles in endosome-based membrane protein targeting in other cell types. We show that EHD1-4 directly associate with ankyrin, provide the first information on the expression and localization of these molecules in primary cardiomyocytes, and demonstrate that EHD1-4 are coexpressed with ankyrin-B in the myocyte perinuclear region. Notably, the expression of multiple EHD proteins is increased in animal models lacking ankyrin-B, and EHD3-deficient cardiomyocytes display aberrant ankyrin-B localization and selective loss of Na/Ca exchanger expression and function. Finally, we report significant modulation of EHD expression following myocardial infarction, suggesting that these proteins may play a key role in regulating membrane excitability in normal and diseased heart. CONCLUSIONS Our findings identify and characterize a new class of cardiac trafficking proteins, define the first group of proteins associated with the ankyrin-based targeting network, and identify potential new targets to modulate membrane excitability in disease. Notably, these data provide the first link between EHD proteins and a human disease model.
Collapse
Affiliation(s)
- Hjalti Gudmundsson
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kieken F, Jović M, Tonelli M, Naslavsky N, Caplan S, Sorgen PL. Structural insight into the interaction of proteins containing NPF, DPF, and GPF motifs with the C-terminal EH-domain of EHD1. Protein Sci 2010; 18:2471-9. [PMID: 19798736 DOI: 10.1002/pro.258] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Eps15 homology (EH)-domain containing proteins are regulators of endocytic membrane trafficking. EH-domain binding to proteins containing the tripeptide NPF has been well characterized, but recent studies have shown that EH-domains are also able to interact with ligands containing DPF or GPF motifs. We demonstrate that the three motifs interact in a similar way with the EH-domain of EHD1, with the NPF motif having the highest affinity due to the presence of an intermolecular hydrogen bond. The weaker affinity for the DPF and GPF motifs suggests that if complex formation occurs in vivo, they may require high ligand concentrations, the presence of successive motifs and/or specific flanking residues.
Collapse
Affiliation(s)
- Fabien Kieken
- Department of Biochemistry and Molecular Biology and Eppley Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198-5870, USA
| | | | | | | | | | | |
Collapse
|
32
|
Heller B, Adu-Gyamfi E, Smith-Kinnaman W, Babbey C, Vora M, Xue Y, Bittman R, Stahelin RV, Wells CD. Amot recognizes a juxtanuclear endocytic recycling compartment via a novel lipid binding domain. J Biol Chem 2010; 285:12308-20. [PMID: 20080965 DOI: 10.1074/jbc.m109.096230] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Polarity proteins promote the asymmetric organization of cells by orienting intracellular sorting mechanisms, such as protein trafficking and cytoskeletal assembly. The localization of individual polarity proteins in turn is often determined by association with factors that mediate contact with other cells or the substratum. This arrangement for the Par and Crb apical polarity complexes at the tight junction is disrupted by the adaptor protein Amot. Amot directly binds the scaffolding proteins Patj and Mupp1 and redistributes them and their binding partners from the plasma membrane to endosomes. However, the mechanism by which Amot is targeted to endosomes is unknown. Here, a novel lipid binding domain within Amot is shown to selectively bind with high affinity to membranes containing monophosphorylated phosphatidylinositols and cholesterol. With similar lipid specificity, Amot inserts into and tubulates membranes in vitro and enlarges perinuclear endosomal compartments in cells. Based on the similar distribution of Amot with cholesterol, Rab11, and Arf6, such membrane interactions are identified at juxtanuclear endocytic recycling compartments. Taken together, these findings indicate that Amot is targeted along with associated apical polarity proteins to the endocytic recycling compartment via this novel membrane binding domain.
Collapse
Affiliation(s)
- Brigitte Heller
- Department of Biochemistry and Molecular Biology, University of Indiana School ofMedicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Cobbe N, Marshall KM, Gururaja Rao S, Chang CW, Di Cara F, Duca E, Vass S, Kassan A, Heck MMS. The conserved metalloprotease invadolysin localizes to the surface of lipid droplets. J Cell Sci 2009; 122:3414-23. [PMID: 19706689 DOI: 10.1242/jcs.044610] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Invadolysin is a metalloprotease conserved in many different organisms, previously shown to be essential in Drosophila with roles in cell division and cell migration. The gene seems to be ubiquitously expressed and four distinct splice variants have been identified in human cells but not in most other species examined. Immunofluorescent detection of human invadolysin in cultured cells reveals the protein to be associated with the surface of lipid droplets. By means of subcellular fractionation, we have independently confirmed the association of invadolysin with lipid droplets. We thus identify invadolysin as the first metalloprotease located on these dynamic organelles. In addition, analysis of larval fat-body morphological appearance and triglyceride levels in the Drosophila invadolysin mutant suggests that invadolysin plays a role in lipid storage or metabolism.
Collapse
Affiliation(s)
- Neville Cobbe
- University of Edinburgh, Queen's Medical Research Institute, Centre for Cardiovascular Science, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Sengupta S, George M, Miller KK, Naik K, Chou J, Cheatham MA, Dallos P, Naramura M, Band H, Zheng J. EHD4 and CDH23 are interacting partners in cochlear hair cells. J Biol Chem 2009; 284:20121-9. [PMID: 19487694 DOI: 10.1074/jbc.m109.025668] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cadherin 23 (CDH23), a transmembrane protein localized near the tips of hair cell stereocilia in the mammalian inner ear, is important for delivering mechanical signals to the mechano-electric transducer channels. To identify CDH23-interacting proteins, a membrane-based yeast two-hybrid screen of an outer hair cell (OHC) cDNA library was performed. EHD4, a member of the C-terminal EH domain containing a protein family involved in endocytic recycling, was identified as a potential interactor. To confirm the interaction, we first demonstrated the EHD4 mRNA expression in hair cells using in situ hybridization. Next, we showed that EHD4 co-localizes and co-immunoprecipitates with CDH23 in mammalian cells. Interestingly, the co-immunoprecipitation was found to be calcium-sensitive. To investigate the role of EHD4 in hearing, compound action potentials were measured in EHD4 knock-out (KO) mice. Although EHD4 KO mice have normal hearing sensitivity, analysis of mouse cochlear lysates revealed a 2-fold increase in EHD1, but no increase in EHD2 or EHD3, in EHD4 KO cochleae compared with wild type, suggesting that a compensatory increase in EHD1 levels may account for the absence of a hearing defect in EHD4 KO mice. Taken together, these data indicate that EHD4 is a novel CDH23-interacting protein that could regulate CDH23 trafficking/localization in a calcium-sensitive manner.
Collapse
Affiliation(s)
- Soma Sengupta
- Department of Communication Sciences and Disorders, Hugh Knowles Center, Northwestern University, Evanston, Illinois 60208, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Martin B, Pearson M, Brenneman R, Golden E, Wood W, Prabhu V, Becker KG, Mattson MP, Maudsley S. Gonadal transcriptome alterations in response to dietary energy intake: sensing the reproductive environment. PLoS One 2009; 4:e4146. [PMID: 19127293 PMCID: PMC2607546 DOI: 10.1371/journal.pone.0004146] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2008] [Accepted: 12/03/2008] [Indexed: 12/21/2022] Open
Abstract
Reproductive capacity and nutritional input are tightly linked and animals' specific responses to alterations in their physical environment and food availability are crucial to ensuring sustainability of that species. We have assessed how alterations in dietary energy intake (both reductions and excess), as well as in food availability, via intermittent fasting (IF), affect the gonadal transcriptome of both male and female rats. Starting at four months of age, male and female rats were subjected to a 20% or 40% caloric restriction (CR) dietary regime, every other day feeding (IF) or a high fat-high glucose (HFG) diet for six months. The transcriptional activity of the gonadal response to these variations in dietary energy intake was assessed at the individual gene level as well as at the parametric functional level. At the individual gene level, the females showed a higher degree of coherency in gonadal gene alterations to CR than the males. The gonadal transcriptional and hormonal response to IF was also significantly different between the male and female rats. The number of genes significantly regulated by IF in male animals was almost 5 times greater than in the females. These IF males also showed the highest testosterone to estrogen ratio in their plasma. Our data show that at the level of gonadal gene responses, the male rats on the IF regime adapt to their environment in a manner that is expected to increase the probability of eventual fertilization of females that the males predict are likely to be sub-fertile due to their perception of a food deficient environment.
Collapse
Affiliation(s)
- Bronwen Martin
- Laboratory of Neurosciences, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Michele Pearson
- Laboratory of Neurosciences, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Randall Brenneman
- Laboratory of Neurosciences, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Erin Golden
- Laboratory of Neurosciences, National Institute on Aging, Baltimore, Maryland, United States of America
| | - William Wood
- Gene Expression and Genomics Unit, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Vinayakumar Prabhu
- Gene Expression and Genomics Unit, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Kevin G. Becker
- Gene Expression and Genomics Unit, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Stuart Maudsley
- Receptor Pharmacology Unit, National Institute on Aging, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
36
|
Abstract
The evolutionarily conserved Eps15 homology domain (EHD)/receptor-mediated endocytosis (RME)-1 family of C-terminal EH domain proteins has recently come under intense scrutiny because of its importance in intracellular membrane transport, especially with regard to the recycling of receptors from endosomes to the plasma membrane. Recent studies have shed new light on the mode by which these adenosine triphosphatases function on endosomal membranes in mammals and Caenorhabditis elegans. This review highlights our current understanding of the physiological roles of these proteins in vivo, discussing conserved features as well as emerging functional differences between individual mammalian paralogs. In addition, these findings are discussed in light of the identification of novel EHD/RME-1 protein and lipid interactions and new structural data for proteins in this family, indicating intriguing similarities to the Dynamin superfamily of large guanosine triphosphatases.
Collapse
Affiliation(s)
- Barth D Grant
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA.
| | | |
Collapse
|
37
|
Bibliography. Current world literature. Atherosclerosis: cell biology and lipoproteins. Curr Opin Lipidol 2008; 19:525-35. [PMID: 18769235 DOI: 10.1097/mol.0b013e328312bffc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
38
|
Abstract
All four of the C-terminal Eps15 homology domain (EHD) proteins have been implicated in the regulation of endocytic trafficking. However, the high level of amino acid sequence identity among these proteins has made it challenging to elucidate the precise function of individual EHD proteins. We demonstrate here with specific peptide antibodies that endogenous EHD4 localizes to Rab5-, early embryonic antigen 1 (EEA1)- and Arf6-containing endosomes and colocalizes with internalized transferrin in the cell periphery. Knock-down of EHD4 expression by both small interfering RNA and short hairpin RNA leads to the generation of enlarged early endosomal structures that contain Rab5 and EEA1 as well as internalized transferrin or major histocompatibility complex class I molecules. In addition, cargo destined for degradation, such as internalized low-density lipoprotein, also accumulates in the enlarged early endosomes in EHD4-depleted cells. Moreover, we have demonstrated that these enlarged early endosomes are enriched in levels of the activated GTP-bound Rab5. Finally, we show that endogenous EHD4 and EHD1 interact in cells, suggesting coordinated involvement in the regulation of receptor transport along the early endosome to endocytic recycling compartment axis. The results presented herein provide evidence that EHD4 is involved in the control of trafficking at the early endosome and regulates exit of cargo toward both the recycling compartment and the late endocytic pathway.
Collapse
Affiliation(s)
- Mahak Sharma
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | | | | |
Collapse
|