1
|
Zhao QG, Ma XL, Xu Q, Song ZT, Bu F, Li K, Han BX, Yan SS, Zhang L, Luo Y, Pei YF. Integrative analysis of transcriptome and proteome wide association studies prioritized functional genes for obesity. Hum Genet 2025; 144:31-41. [PMID: 39495296 DOI: 10.1007/s00439-024-02714-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Genome-wide association studies have identified dozens of genomic loci for obesity. However, functional genes and their detailed genetic mechanisms underlying these loci are mainly unknown. In this study, we conducted an integrative study to prioritize plausibly functional genes by combining information from genome-, transcriptome- and proteome-wide association analyses. METHODS We first conducted proteome-wide association analyses and transcriptome-wide association analyses for the six obesity-related traits. We then performed colocalization analysis on the identified loci shared between the proteome- and transcriptome-association analyses. Finally, we validated the identified genes with other plasma/blood reference panels. The highlighted genes were assessed for expression of other tissues, single-cell and tissue specificity, and druggability. RESULTS We prioritized 4 high-confidence genes (FASN, ICAM1, PDCD6IP, and YWHAB) by proteome-wide association studies, transcriptome-wide association studies, and colocalization analyses, which consistently influenced the variation of obesity traits at both mRNA and protein levels. These 4 genes were successfully validated using other plasma/blood reference panels. These 4 genes shared regulatory structures in obesity-related tissues. Single-cell and tissue-specific analyses showed that FASN and ICAM1 were explicitly expressed in metabolism- and immunity-related tissues and cells. Furthermore, FASN and ICAM1 had been developed as drug targets. CONCLUSION Our study provided novel promising protein targets for further mechanistic and therapeutic studies of obesity.
Collapse
Affiliation(s)
- Qi-Gang Zhao
- Department of Orthopedics, Taicang Affiliated Hospital of Soochow University, 58 Changsheng Rd., Suzhou Taicang City, 215400, Jiangsu Province, PR China
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, 199 Ren-ai Rd., Suzhou City, 215123, Jiangsu Province, PR China
| | - Xin-Ling Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, 199 Ren-ai Rd., Suzhou City, 215123, Jiangsu Province, PR China
| | - Qian Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, 199 Ren-ai Rd., Suzhou City, 215123, Jiangsu Province, PR China
| | - Zi-Tong Song
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, 199 Ren-ai Rd., Suzhou City, 215123, Jiangsu Province, PR China
| | - Fan Bu
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, 199 Ren-ai Rd., Suzhou City, 215123, Jiangsu Province, PR China
| | - Kuan Li
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, 199 Ren-ai Rd., Suzhou City, 215123, Jiangsu Province, PR China
| | - Bai-Xue Han
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, 199 Ren-ai Rd., Suzhou City, 215123, Jiangsu Province, PR China
| | - Shan-Shan Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, 199 Ren-ai Rd., Suzhou City, 215123, Jiangsu Province, PR China
| | - Lei Zhang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou City, Jiangsu, PR China
| | - Yuan Luo
- Department of Orthopedics, Taicang Affiliated Hospital of Soochow University, 58 Changsheng Rd., Suzhou Taicang City, 215400, Jiangsu Province, PR China.
| | - Yu-Fang Pei
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, 199 Ren-ai Rd., Suzhou City, 215123, Jiangsu Province, PR China.
| |
Collapse
|
2
|
Koch RL, Stanton JB, McClatchy S, Churchill GA, Craig SW, Williams DN, Johns ME, Chase KR, Thiesfeldt DL, Flynt JC, Pazdro R. Discovery of genomic loci for liver health and steatosis reveals overlap with glutathione redox genetics. Redox Biol 2024; 75:103248. [PMID: 38917671 PMCID: PMC11254179 DOI: 10.1016/j.redox.2024.103248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/27/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver condition in the United States, encompassing a wide spectrum of liver pathologies including steatosis, steatohepatitis, fibrosis, and cirrhosis. Despite its high prevalence, there are no medications currently approved by the Food and Drug Administration for the treatment of NAFLD. Recent work has suggested that NAFLD has a strong genetic component and identifying causative genes will improve our understanding of the molecular mechanisms contributing to NAFLD and yield targets for future therapeutic investigations. Oxidative stress is known to play an important role in NAFLD pathogenesis, yet the underlying mechanisms accounting for disturbances in redox status are not entirely understood. To better understand the relationship between the glutathione redox system and signs of NAFLD in a genetically-diverse population, we measured liver weight, serum biomarkers aspartate aminotransferase (AST) and alanine aminotransferase (ALT), and graded liver pathology in a large cohort of Diversity Outbred mice. We compared hepatic endpoints to those of the glutathione redox system previously measured in the livers and kidneys of the same mice, and we screened for statistical and genetic associations using the R/qtl2 software. We discovered several novel genetic loci associated with markers of liver health, including loci that were associated with both liver steatosis and glutathione redox status. Candidate genes within each locus point to possible new mechanisms underlying the complex relationship between NAFLD and the glutathione redox system, which could have translational implications for future studies targeting NAFLD pathology.
Collapse
Affiliation(s)
- Rebecca L Koch
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA, 30602
| | - James B Stanton
- Department of Pathology, University of Georgia, Athens, GA, USA, 30602
| | | | | | - Steven W Craig
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA, 30602
| | - Darian N Williams
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA, 30602
| | - Mallory E Johns
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA, 30602
| | - Kylah R Chase
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA, 30602
| | - Dana L Thiesfeldt
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA, 30602
| | - Jessica C Flynt
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA, 30602
| | - Robert Pazdro
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA, 30602.
| |
Collapse
|
3
|
Metabolomic Analysis of Serum and Tear Samples from Patients with Obesity and Type 2 Diabetes Mellitus. Int J Mol Sci 2022; 23:ijms23094534. [PMID: 35562924 PMCID: PMC9105607 DOI: 10.3390/ijms23094534] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 12/14/2022] Open
Abstract
Metabolomics strategies are widely used to examine obesity and type 2 diabetes (T2D). Patients with obesity (n = 31) or T2D (n = 26) and sex- and age-matched controls (n = 28) were recruited, and serum and tear samples were collected. The concentration of 23 amino acids and 10 biogenic amines in serum and tear samples was analyzed. Statistical analysis and Pearson correlation analysis along with network analysis were carried out. Compared to controls, changes in the level of 6 analytes in the obese group and of 10 analytes in the T2D group were statistically significant. For obesity, the energy generation, while for T2D, the involvement of NO synthesis and its relation to insulin signaling and inflammation, were characteristic. We found that BCAA and glutamine metabolism, urea cycle, and beta-oxidation make up crucial parts of the metabolic changes in T2D. According to our data, the retromer-mediated retrograde transport, the ethanolamine metabolism, and, consequently, the endocannabinoid signaling and phospholipid metabolism were characteristic of both conditions and can be relevant pathways to understanding and treating insulin resistance. By providing potential therapeutic targets and new starting points for mechanistic studies, our results emphasize the importance of complex data analysis procedures to better understand the pathomechanism of obesity and diabetes.
Collapse
|
4
|
Silva-Gaona OG, Guzmán-Flores JM, Hernández-Ortiz M, Vargas-Ortiz K, Ramírez-Emiliano J, Encarnación-Guevara S, Pérez-Vázquez V. Curcumin Reverts the Protein Differential Expression in the Liver of the Diabetic Obese db/db Mice. CURR PROTEOMICS 2022. [DOI: 10.2174/1570164618666210114112642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
In type 2 diabetic mouse liver, hyperglycemia, and insulin modify gene expression. Curcumin is a
powerful antioxidant and antidiabetic agent that regulates the gene expression of different signaling pathways through
various transcription factors. Therefore, we hypothesized that curcumin modifies the protein expression profile in the liver
of diabetic db/db mice.
Objective:
To determine the effects of curcumin on the liver protein profile of diabetic db/db mice.
Methods:
db/db and wild type (WT) male mice were allocated in four groups, and they were fed for eight weeks. Three WT
and three diabetic db/db mice received a standard diet (SD; WT and db/db groups, respectively); three WT and three
diabetic db/db mice received a SD supplemented with 0.75 % (w/w) curcumin (WT+C and db/db+C groups, respectively).
Liver proteins were separated by 2D electrophoresis. Differential protein expression analysis was performed on
ImageMaster 2D Platinum software, and selected proteins were identified by MALDI-TOF-MS and subjected to enrichment
analysis using STRING and DAVID databases.
Results:
Thirty-six proteins with differential expression due to the diabetic background and curcumin treatment were found;
these proteins participate in the metabolism of amino acids, carbohydrates, and lipids. Interestingly, the altered expression of
seven proteins was prevented in the liver of the diabetic mice that received curcumin.
Conclusions:
Among all differentially expressed proteins, curcumin reverted the altered expression of seven proteins. Thus,
although it was observed that curcumin did not affect the biochemical parameters, it does modify the expression of some
liver proteins in diabetic mice.
Collapse
Affiliation(s)
- Oscar Gerardo Silva-Gaona
- Dpto. de Ciencias Médicas, División de Ciencias de la Salud, Campus León, Universidad de Guanajuato, León, Guanajuato., México
| | - Juan Manuel Guzmán-Flores
- Depto. de Salud, División de Ciencias Biomédicas, Centro Universitario de los Altos, Universidad
de Guadalajara, Tepatitlán, Jalisco, México
| | | | - Katya Vargas-Ortiz
- Dpto. de Ciencias Médicas, División de Ciencias de la Salud, Campus León, Universidad de Guanajuato, León, Guanajuato., México
| | - Joel Ramírez-Emiliano
- Dpto. de Ciencias Médicas, División de Ciencias de la Salud, Campus León, Universidad de Guanajuato, León, Guanajuato., México
| | - Sergio Encarnación-Guevara
- Dpto. de Ciencias Médicas, División de Ciencias de la Salud, Campus León, Universidad de Guanajuato, León, Guanajuato., México
| | - Victoriano Pérez-Vázquez
- Dpto. de Ciencias Médicas, División de Ciencias de la Salud, Campus León, Universidad de Guanajuato, León, Guanajuato., México
| |
Collapse
|
5
|
Miranda-Nuñez JE, Zamilpa-Alvarez A, Fortis-Barrera A, Alarcon-Aguilar FJ, Loza-Rodriguez H, Gomez-Quiroz LE, Salas-Silva S, Flores-Cruz M, Zavala-Sanchez MA, Blancas-Flores G. GLUT4 translocation in C2C12 myoblasts and primary mouse hepatocytes by an antihyperglycemic flavone from Tillandsia usneoides. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 89:153622. [PMID: 34161895 DOI: 10.1016/j.phymed.2021.153622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/23/2021] [Accepted: 05/30/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Type 2 Diabetes (T2D) is characterized by deregulation in carbohydrate and lipid metabolism, with a very high mortality rate. Glucose Transporter type 4 (GLUT4) plays a crucial role in T2D and represents a therapeutic target of interest. Tillandsia usneoides (T. usneoides) is a plant used as a remedy for diabetes. T. usneoides decreased blood glucose in different experimental models. However, the involvement of GLUT4 in this effect has not yet been explored. PURPOSE This study aimed to investigate whether any component in T. usneoides might participate in the effect on blood glucose through a bioassay-guided fractionation, testing its potential antihyperglycemic effect in mice, as well as its influence on GLUT4 translocation in C2C12 myoblasts and primary hepatocytes. METHODS The aqueous extract and the Ethyl Acetate fraction (TU-AcOEt) of T. usneoides were evaluated in a hypoglycemic activity bioassay and in the glucose tolerance test in CD-1 mice. TU-AcOEt was fractionated, obtaining five fractions that were studied in an additional glucose tolerance test. C1F3 was fractioned again, and its fractions (C2F9-12, C2F22-25, and C2F38-44) were examined by HPLC. The C2F38-44 fraction was analyzed by Mass Spectrometry (MS) and subjected to additional fractionation. The fraction C3F6-9 was explored by Nuclear Magnetic Resonance (NMR), resulting in 5,7,4´-trihydroxy-3,6,3´,5´-tetramethoxyflavone (Flav1). Subsequently, a viability test was performed to evaluate the cytotoxic effect of Flav1 and fractions C2F9-12, C2F22-25. C2F38-44, and C3F30-41 in C2C12 myoblasts and primary mouse hepatocytes. Confocal microscopy was also performed to assess the effect of Flav1 and fractions on GLUT4 translocation. RESULTS The TU-AcOEt fraction exhibited a hypoglycemic and antihyperglycemic effect in mice, and its fractionation resulted in five fractions, among which fraction C1F3 decreased blood glucose. MS and NMR analysis revealed the presence of Flav1. Finally, Flav1 significantly promoted the translocation of GLUT4 in C2C12 myoblasts and primary hepatocytes. CONCLUSION To date, Flav1 has not been reported to have activity in GLUT4; this study provides evidence that T. usneoides is a plant with the potential to develop novel therapeutic agents for the control of T2D.
Collapse
Affiliation(s)
| | - Alejandro Zamilpa-Alvarez
- Departamento de Fitoquímica Farmacológica, Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social, Xochitepec, Morelos, México
| | - Angeles Fortis-Barrera
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Ciudad de México, México
| | - Francisco Javier Alarcon-Aguilar
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Ciudad de México, México
| | - Hilda Loza-Rodriguez
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Ciudad de México, México
| | - Luis E Gomez-Quiroz
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Ciudad de México, México
| | - Soraya Salas-Silva
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Ciudad de México, México
| | - Maria Flores-Cruz
- Departamento el Hombre y su Ambiente, DCBS, Universidad Autónoma Metropolitana-Xochimilco, Calzada del Hueso 1100, Ciudad de México, México
| | - Miguel Angel Zavala-Sanchez
- Departamento de Sistemas Biológicos, DCBS, Universidad Autónoma Metropolitana-Xochimilco, Calzada del Hueso 1100, Ciudad de México, México
| | - Gerardo Blancas-Flores
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Ciudad de México, México.
| |
Collapse
|
6
|
Ji L, Wang Q, Liu M, Zhu C, Xiao Y, Han J, Fang Y, Ye J, Yin J, Wei L. The 14-3-3 protein YWHAB inhibits glucagon-induced hepatic gluconeogenesis through interacting with the glucagon receptor and FOXO1. FEBS Lett 2021; 595:1275-1288. [PMID: 33641163 DOI: 10.1002/1873-3468.14063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 01/15/2021] [Accepted: 01/23/2021] [Indexed: 01/09/2023]
Abstract
Glucagon antagonism has been reported as a new therapeutic approach to hyperglycaemia. As the 14-3-3 protein YWHAB has been identified as a regulator of the glucagon receptor (GCGR) by affinity purification and mass spectrometry, we examined the role of YWHAB in vivo. Ywhab knockout mice display impaired blood glucose homeostasis only under pyruvate stimulation. Deletion of Ywhab in mouse primary hepatocytes (MPHs) increases hepatocyte glucose production by magnifying the effect of glucagon. Mechanistic analysis indicates that YWHAB forms a phosphorylation-dependent complex with GCGR and directly interacts with forkhead box O1 (FOXO1). Together, these results reveal the inhibitory role of YWHAB in glucagon-mediated hepatic glucose production, which may be a potential target for the control of gluconeogenesis and associated metabolic diseases.
Collapse
Affiliation(s)
- Linlin Ji
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Qianqian Wang
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
- Department of Endocrinology, School of Medicine, Shanghai Tongren Hospital, affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Mengdan Liu
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Chaoyu Zhu
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Yuanyuan Xiao
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Junfeng Han
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Yunyun Fang
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Jianping Ye
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Jun Yin
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Li Wei
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| |
Collapse
|
7
|
Integrative Analyses of Genes Associated with Fulminant Type 1 Diabetes. J Immunol Res 2020; 2020:1025857. [PMID: 33083497 PMCID: PMC7559223 DOI: 10.1155/2020/1025857] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 08/25/2020] [Accepted: 09/15/2020] [Indexed: 12/21/2022] Open
Abstract
Objective Fulminant type 1 diabetes (FT1D) is a type of type 1 diabetes, which is characterized by rapid onset of disease and severe metabolic disorders. We intend to screen for crucial genes and potential molecular mechanisms in FT1D in this study. Method We downloaded GSE44314, which includes six healthy controls and five patients with FT1D, from the GEO database. Identification of differentially expressed genes (DEGs) was performed by NetworkAnalyst. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses of DEGs were screened by an online tool-Database for Annotation, Visualization, and Integration Discovery (DAVID). Protein-protein interaction (PPI) network and hub genes among DEGs were analyzed by NetworkAnalyst. And we also use NetworkAnalyst to find out the microRNAs (miRNAs) and transcription factors (TFs) which regulate the expression of DEGs. Result We identified 130 DEGs (60 upregulated and 70 downregulated DEGs) between healthy controls and FT1D patients. GO analysis results revealed that DEGs were mostly enriched in generation of precursor metabolites and energy, neurohypophyseal hormone activity, and mitochondrial inner membrane. KEGG pathway analysis demonstrated that DEGs were mostly involved in nonalcoholic fatty liver disease. Results indicated that NCOA1, SRF, ERBB3, EST1, TOP1, UBE2S, INO80, COX7C, ITGAV, and COX6C were the top hub genes in the PPI network. Furthermore, we recognized that LDLR, POTEM, IFNAR2, BAZ2A, and SRF were the top hub genes in the miRNA-target gene network, and SRF, TSPAN4, CD59, ETS1, and SLC25A25 were the top hub genes in the TF-target gene network. Conclusion Our study pinpoints key genes and pathways associated with FT1D by a sequence of bioinformatics analysis on DEGs. These identified genes and pathways provide more detailed molecular mechanisms of FT1D and may provide novel therapeutic targets.
Collapse
|
8
|
Yang N, Yu L, Deng Y, Han Q, Wang J, Yu L, Zhai Z, Li W. Identification and characterization of proteins that are differentially expressed in adipose tissue of olanzapine-induced insulin resistance rat by iTRAQ quantitative proteomics. J Proteomics 2019; 212:103570. [PMID: 31706944 DOI: 10.1016/j.jprot.2019.103570] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 10/22/2019] [Accepted: 10/30/2019] [Indexed: 12/31/2022]
Abstract
Olanzapine is commonly used to treat schizophrenia. However, long-term administration of olanzapine causes metabolic side effects, such as insulin resistance (IR), which seriously affects patients' quality of life. Both diagnostic and prognostic markers are urgently needed to increase patient compliance. We applied isobaric tags for relative and absolute quantitation (iTRAQ) labeling combined with 2D LC/MS/MS technology to identify the differentially expressed proteins in olanzapine-induced IR rats. A total of 3194 proteins were identified from rat adipose tissues, and 270 differentially expressed proteins were screened out with a ratio threshold >1.5-fold or <0.67-fold. Based on a bioinformatics analysis and literature search, we selected six candidates (MYH1, MYL2, Cp, FABP4, apoA-IV, and Ywhaz) from a set of 270 proteins and verified these proteins by western blot; the expression of these proteins coincided with the LC-MS/MS results. Finally, the biological roles of FABP4 and apoA-IV, which are two novel IR-related proteins identified in the present study, were verified in 3T3-L1 cells. These data suggest that these two proteins acted on olanzapine-induced IR via the IRS-1/AKT signaling pathway. Our results provide a dataset of potential targets to explore the mechanism in olanzapine-induced IR and reveal the new roles of FABP4 and apoA-IV in olanzapine-induced IR. SIGNIFICANCE: The proteomic analysis of this study revealed the target associated with olanzapine-induced IR and provided relevant insights into the molecular functions, biological processes, and signaling pathways in these targets. Protein MYH1, MYL2, Cp, FABP4, apoA-IV, and Ywhaz may be potential biomarkers, and protein FABP4 and apoA-IV were considered as promising targets in olanzapineinduced IR. Therefore, if the performance of the proposed biomarkers is further confirmed, these proteins can provide powerful targets for exploring the mechanism of olanzapine-induced IR.
Collapse
Affiliation(s)
- Ni Yang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liangyu Yu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yahui Deng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qiangqiang Han
- Building B5, Biolake, East Lake New Technology Development Zone, Wuhan, China
| | - Jing Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lixiu Yu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhongfang Zhai
- Shanghai City shanghai general hospital, No. 650 Xinsongjiang Road, Songjiang District, Shanghai, China
| | - Weiyong Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
9
|
Ding J, Zhu YT, Yang L, Tang J, Wang YY, Chen Y, Cheng K, Liu JQ, Zhang YN, Li ZK, Du Y, Qiu M, Liu JY. 14-3-3zeta is involved in the anticancer effect of metformin in colorectal carcinoma. Carcinogenesis 2018; 39:493-502. [PMID: 29390122 DOI: 10.1093/carcin/bgy008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Metformin is a promising drug for cancer prevention and treatment, especially in the diabetic population. We aimed to test whether 14-3-3zeta affects the anticancer effect of metformin on colorectal carcinoma (CRC). In this study, we confirmed that higher 14-3-3zeta expression was found in CRC tissues than in pericarcinoma tissues, and in CRC tissue of patients with diabetes than in those without diabetes. A knockdown of 14-3-3zeta inhibited CRC proliferation and promoted apoptosis in vitro and in vivo. Then, we created stable cell lines with under-expressed 14-3-3zeta from SW480 and HCT15 cells after infection by a lentiviral vector carrying short hairpin RNA targeting 14-3-3zeta (named LV-sh14-3-3zeta). Of note, metformin induced apoptosis and retarded tumor growth in the CRCs with overexpressed 14-3-3zeta, whereas this action was attenuated when 14-3-3zeta was knocked down. Moreover, either metformin or downregulation of 14-3-3zeta noticeably activated AMP-dependent protein kinase (AMPK) signaling, whereas the effect of metformin was attenuated when the 14-3-3zeta expression was decreased. Taken together, our results suggest that 14-3-3zeta may be associated with carcinogenesis and poor prognosis of CRCs associated with diabetes, and metformin may reverse the AMPK inhibition caused by 14-3-3zeta in CRCs in the background of diabetes. Our study should lead to a better understanding of the anticancer activity of metformin and points to possible application of metformin to the treatment of cancers overexpressing 14-3-3zeta.
Collapse
Affiliation(s)
- Jing Ding
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
- Department of Oncology, Cancer Hospital Affiliate to School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yue-Ting Zhu
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Lie Yang
- Department of Gastrointestinal Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Jie Tang
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Yu-Yi Wang
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Ye Chen
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Ke Cheng
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Jia-Qi Liu
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Yun-Ni Zhang
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhi-Ke Li
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Yang Du
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Meng Qiu
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Ji-Yan Liu
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
10
|
Lohr K, Pachl F, Moghaddas Gholami A, Geillinger KE, Daniel H, Kuster B, Klingenspor M. Reduced mitochondrial mass and function add to age-related susceptibility toward diet-induced fatty liver in C57BL/6J mice. Physiol Rep 2017; 4:4/19/e12988. [PMID: 27694529 PMCID: PMC5064140 DOI: 10.14814/phy2.12988] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/09/2016] [Indexed: 01/11/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a major health burden in the aging society with an urging medical need for a better understanding of the underlying mechanisms. Mitochondrial fatty acid oxidation and mitochondrial‐derived reactive oxygen species (ROS) are considered critical in the development of hepatic steatosis, the hallmark of NAFLD. Our study addressed in C57BL/6J mice the effect of high fat diet feeding and age on liver mitochondria at an early stage of NAFLD development. We therefore analyzed functional characteristics of hepatic mitochondria and associated alterations in the mitochondrial proteome in response to high fat feeding in adolescent, young adult, and middle‐aged mice. Susceptibility to diet‐induced obesity increased with age. Young adult and middle‐aged mice developed fatty liver, but not adolescent mice. Fat accumulation was negatively correlated with an age‐related reduction in mitochondrial mass and aggravated by a reduced capacity of fatty acid oxidation in high fat‐fed mice. Irrespective of age, high fat diet increased ROS production in hepatic mitochondria associated with a balanced nuclear factor erythroid‐derived 2 like 2 (NFE2L2) dependent antioxidative response, most likely triggered by reduced tethering of NFE2L2 to mitochondrial phosphoglycerate mutase 5. Age indirectly influenced mitochondrial function by reducing mitochondrial mass, thus exacerbating diet‐induced fat accumulation. Therefore, consideration of age in metabolic studies must be emphasized.
Collapse
Affiliation(s)
- Kerstin Lohr
- Chair of Molecular Nutritional Medicine, Technische Universität München, Else Kröner Fresenius Center for Nutritional Medicine, Freising-Weihenstephan, Germany Z I E L - Research Center for Nutrition and Food Sciences, Technische Universität München, Freising-Weihenstephan, Germany
| | - Fiona Pachl
- Chair of Proteomics and Bioanalytics, Technische Universität München Bavarian Biomolecular Mass Spectrometry Center, Freising-Weihenstephan, Germany
| | - Amin Moghaddas Gholami
- Chair of Proteomics and Bioanalytics, Technische Universität München Bavarian Biomolecular Mass Spectrometry Center, Freising-Weihenstephan, Germany
| | - Kerstin E Geillinger
- Z I E L - Research Center for Nutrition and Food Sciences, Technische Universität München, Freising-Weihenstephan, Germany Nutritional Physiology, Technische Universität München, Freising-Weihenstephan, Germany
| | - Hannelore Daniel
- Z I E L - Research Center for Nutrition and Food Sciences, Technische Universität München, Freising-Weihenstephan, Germany Nutritional Physiology, Technische Universität München, Freising-Weihenstephan, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technische Universität München Bavarian Biomolecular Mass Spectrometry Center, Freising-Weihenstephan, Germany
| | - Martin Klingenspor
- Chair of Molecular Nutritional Medicine, Technische Universität München, Else Kröner Fresenius Center for Nutritional Medicine, Freising-Weihenstephan, Germany Z I E L - Research Center for Nutrition and Food Sciences, Technische Universität München, Freising-Weihenstephan, Germany
| |
Collapse
|
11
|
Ke M, Wu H, Zhu Z, Zhang C, Zhang Y, Deng Y. Differential proteomic analysis of white adipose tissues from T2D KKAy mice by LC-ESI-QTOF. Proteomics 2017; 17. [PMID: 27995753 DOI: 10.1002/pmic.201600219] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 11/28/2016] [Accepted: 12/13/2016] [Indexed: 12/14/2022]
Abstract
Type 2 diabetes (T2D) has become a worldwide increasingly social health burden for its high morbidity and heightened prevalence. As one of the main tissues involved in uptake of glucose under the stimulation of insulin, WAT plays very important role in metabolic and homeostasis regulation. We performed a differential proteomics study to investigate alterations in epididymis fat pad of high fat diet fed T2D KKAy mice compared to normal fed C57BL/6J mice, by 18 O-labeling relative quantitative technique. Among 329 confidently identified proteins, 121 proteins showed significant changes with CV ≤ 20% (fold changes of >2 or <0.5 as threshold). According to GO classification, we found that altered proteins contained members of biological processes of metabolic process, oxidative stress, ion homeostasis, apoptosis and cell division. In metabolic, proteins assigned to fatty acid biosynthesis (FAS etc.) were decreased, the key enzyme (ACOX3) in β-oxidation process was increased. Increased glycolysis enzymes (ENOB etc.) and decreased TCA cycle related enzymes (SCOT1 etc.) suggested that glucose metabolism in mitochondria of T2D mice might be impaired. Elevated oxidative stress was observed with alterations of a series of oxidordeuctase (QSOX1 etc.). Besides, alterations of ion homeostasis (AT2C1 etc.) proteins were also observed. The enhancement of cell proliferation associated proteins (ELYS etc.) and inhibition of apoptosis associated proteins (RASF6 etc.) in WAT might contributed to the fat pad and body weight gain. Overall, these changes in WAT may serve as a reference for understanding the functional mechanism of T2D.
Collapse
Affiliation(s)
- Ming Ke
- Beijing Institute of Technology, School of life science, Haidian, Beijing, P. R. China
| | - Hanyan Wu
- Beijing Institute of Technology, School of life science, Haidian, Beijing, P. R. China
| | - Zhaoyang Zhu
- Beijing Institute of Technology, School of life science, Haidian, Beijing, P. R. China
| | - Chi Zhang
- Beijing Institute of Technology, School of life science, Haidian, Beijing, P. R. China
| | - Yongqian Zhang
- Beijing Institute of Technology, School of life science, Haidian, Beijing, P. R. China
| | - Yunlin Deng
- Beijing Institute of Technology, School of life science, Haidian, Beijing, P. R. China
| |
Collapse
|
12
|
Genome-wide association study for feed efficiency and growth traits in U.S. beef cattle. BMC Genomics 2017; 18:386. [PMID: 28521758 PMCID: PMC5437562 DOI: 10.1186/s12864-017-3754-y] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 05/03/2017] [Indexed: 11/13/2022] Open
Abstract
Background Single nucleotide polymorphism (SNP) arrays for domestic cattle have catalyzed the identification of genetic markers associated with complex traits for inclusion in modern breeding and selection programs. Using actual and imputed Illumina 778K genotypes for 3887 U.S. beef cattle from 3 populations (Angus, Hereford, SimAngus), we performed genome-wide association analyses for feed efficiency and growth traits including average daily gain (ADG), dry matter intake (DMI), mid-test metabolic weight (MMWT), and residual feed intake (RFI), with marker-based heritability estimates produced for all traits and populations. Results Moderate and/or large-effect QTL were detected for all traits in all populations, as jointly defined by the estimated proportion of variance explained (PVE) by marker effects (PVE ≥ 1.0%) and a nominal P-value threshold (P ≤ 5e-05). Lead SNPs with PVE ≥ 2.0% were considered putative evidence of large-effect QTL (n = 52), whereas those with PVE ≥ 1.0% but < 2.0% were considered putative evidence for moderate-effect QTL (n = 35). Identical or proximal lead SNPs associated with ADG, DMI, MMWT, and RFI collectively supported the potential for either pleiotropic QTL, or independent but proximal causal mutations for multiple traits within and between the analyzed populations. Marker-based heritability estimates for all investigated traits ranged from 0.18 to 0.60 using 778K genotypes, or from 0.17 to 0.57 using 50K genotypes (reduced from Illumina 778K HD to Illumina Bovine SNP50). An investigation to determine if QTL detected by 778K analysis could also be detected using 50K genotypes produced variable results, suggesting that 50K analyses were generally insufficient for QTL detection in these populations, and that relevant breeding or selection programs should be based on higher density analyses (imputed or directly ascertained). Conclusions Fourteen moderate to large-effect QTL regions which ranged from being physically proximal (lead SNPs ≤ 3Mb) to fully overlapping for RFI, DMI, ADG, and MMWT were detected within and between populations, and included evidence for pleiotropy, proximal but independent causal mutations, and multi-breed QTL. Bovine positional candidate genes for these traits were functionally conserved across vertebrate species. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3754-y) contains supplementary material, which is available to authorized users.
Collapse
|
13
|
Pedersen BA, Wang W, Taylor JF, Khattab OS, Chen YH, Edwards RA, Yazdi PG, Wang PH. Hepatic proteomic analysis revealed altered metabolic pathways in insulin resistant Akt1(+/-)/Akt2(-/-) mice. Metabolism 2015; 64:1694-703. [PMID: 26455965 PMCID: PMC4641788 DOI: 10.1016/j.metabol.2015.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 08/19/2015] [Accepted: 09/08/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE The aim of this study was to identify liver proteome changes in a mouse model of severe insulin resistance and markedly decreased leptin levels. METHODS Two-dimensional differential gel electrophoresis was utilized to identify liver proteome changes in AKT1(+/-)/AKT2(-/-) mice. Proteins with altered levels were identified with tandem mass spectrometry. Ingenuity Pathway Analysis was performed for the interpretation of the biological significance of the observed proteomic changes. RESULTS 11 proteins were identified from 2 biological replicates to be differentially expressed by a ratio of at least 1.3 between age-matched insulin resistant (Akt1(+/-)/Akt2(-/-)) and wild type mice. Albumin and mitochondrial ornithine aminotransferase were detected from multiple spots, which suggest post-translational modifications. Enzymes of the urea cycle were common members of top regulated pathways. CONCLUSION Our results help to unveil the regulation of the liver proteome underlying altered metabolism in an animal model of severe insulin resistance.
Collapse
Affiliation(s)
- Brian A Pedersen
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA
- Department of Medicine, University of California at Irvine, Irvine, CA 92697, USA
| | - Weiwen Wang
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA
- Department of Medicine, University of California at Irvine, Irvine, CA 92697, USA
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami School of Medicine, Miami, FL, 33136
| | - Jared F Taylor
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA
- Department of Medicine, University of California at Irvine, Irvine, CA 92697, USA
| | - Omar S Khattab
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA
| | - Yu-Han Chen
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA
- Department of Physiology & Biophysics, University of California at Irvine, Irvine, CA 92697, USA
| | - Robert A Edwards
- Department of Pathology, University of California at Irvine, Irvine, CA 92697, USA
| | - Puya G Yazdi
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA
- Department of Medicine, University of California at Irvine, Irvine, CA 92697, USA
| | - Ping H Wang
- UC Irvine Diabetes Center, University of California at Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California at Irvine, Irvine, CA 92697, USA
- Department of Medicine, University of California at Irvine, Irvine, CA 92697, USA
- Department of Biological Chemistry, University of California at Irvine, Irvine, CA 92697, USA
- Department of Physiology & Biophysics, University of California at Irvine, Irvine, CA 92697, USA
| |
Collapse
|
14
|
Liu Y, Yan G, Gao M, Deng C, Zhang X. Membrane protein isolation and identification by covalent binding for proteome research. Proteomics 2015; 15:3892-900. [DOI: 10.1002/pmic.201400572] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 08/21/2015] [Accepted: 09/11/2015] [Indexed: 02/02/2023]
Affiliation(s)
- Yiying Liu
- Department of Chemistry, Institutes of Biomedical Sciences; Fudan University; Shanghai P. R. China
| | - Guoquan Yan
- Department of Chemistry, Institutes of Biomedical Sciences; Fudan University; Shanghai P. R. China
| | - Mingxia Gao
- Department of Chemistry, Institutes of Biomedical Sciences; Fudan University; Shanghai P. R. China
| | - Chunhui Deng
- Department of Chemistry, Institutes of Biomedical Sciences; Fudan University; Shanghai P. R. China
| | - Xiangmin Zhang
- Department of Chemistry, Institutes of Biomedical Sciences; Fudan University; Shanghai P. R. China
| |
Collapse
|
15
|
Willebrords J, Pereira IVA, Maes M, Crespo Yanguas S, Colle I, Van Den Bossche B, Da Silva TC, de Oliveira CPMS, Andraus W, Alves VA, Cogliati B, Vinken M. Strategies, models and biomarkers in experimental non-alcoholic fatty liver disease research. Prog Lipid Res 2015; 59:106-125. [PMID: 26073454 PMCID: PMC4596006 DOI: 10.1016/j.plipres.2015.05.002] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/13/2015] [Accepted: 05/13/2015] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease encompasses a spectrum of liver diseases, including simple steatosis, steatohepatitis, liver fibrosis and cirrhosis and hepatocellular carcinoma. Non-alcoholic fatty liver disease is currently the most dominant chronic liver disease in Western countries due to the fact that hepatic steatosis is associated with insulin resistance, type 2 diabetes mellitus, obesity, metabolic syndrome and drug-induced injury. A variety of chemicals, mainly drugs, and diets is known to cause hepatic steatosis in humans and rodents. Experimental non-alcoholic fatty liver disease models rely on the application of a diet or the administration of drugs to laboratory animals or the exposure of hepatic cell lines to these drugs. More recently, genetically modified rodents or zebrafish have been introduced as non-alcoholic fatty liver disease models. Considerable interest now lies in the discovery and development of novel non-invasive biomarkers of non-alcoholic fatty liver disease, with specific focus on hepatic steatosis. Experimental diagnostic biomarkers of non-alcoholic fatty liver disease, such as (epi)genetic parameters and '-omics'-based read-outs are still in their infancy, but show great promise. In this paper, the array of tools and models for the study of liver steatosis is discussed. Furthermore, the current state-of-art regarding experimental biomarkers such as epigenetic, genetic, transcriptomic, proteomic and metabonomic biomarkers will be reviewed.
Collapse
Affiliation(s)
- Joost Willebrords
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Isabel Veloso Alves Pereira
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, São Paulo, Brazil.
| | - Michaël Maes
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Sara Crespo Yanguas
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Isabelle Colle
- Department of Hepatology and Gastroenterology, Algemeen Stedelijk Ziekenhuis Campus Aalst, Merestraat 80, 9300 Aalst, Belgium.
| | - Bert Van Den Bossche
- Department of Abdominal Surgery and Hepato-Pancreatico-Biliary Surgery, Algemeen Stedelijk Ziekenhuis Campus Aalst, Merestraat 80, 9300 Aalst, Belgium.
| | - Tereza Cristina Da Silva
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, São Paulo, Brazil.
| | | | - Wellington Andraus
- Department of Gastroenterology, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, São Paulo, Brazil.
| | - Venâncio Avancini Alves
- Laboratory of Medical Investigation, Department of Pathology, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, São Paulo, Brazil.
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, São Paulo, Brazil.
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| |
Collapse
|
16
|
Han J, Zhang M, Froese S, Dai FF, Robitaille M, Bhattacharjee A, Huang X, Jia W, Angers S, Wheeler MB, Wei L. The Identification of Novel Protein-Protein Interactions in Liver that Affect Glucagon Receptor Activity. PLoS One 2015; 10:e0129226. [PMID: 26075596 PMCID: PMC4468146 DOI: 10.1371/journal.pone.0129226] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 05/06/2015] [Indexed: 11/18/2022] Open
Abstract
Glucagon regulates glucose homeostasis by controlling glycogenolysis and gluconeogenesis in the liver. Exaggerated and dysregulated glucagon secretion can exacerbate hyperglycemia contributing to type 2 diabetes (T2D). Thus, it is important to understand how glucagon receptor (GCGR) activity and signaling is controlled in hepatocytes. To better understand this, we sought to identify proteins that interact with the GCGR to affect ligand-dependent receptor activation. A Flag-tagged human GCGR was recombinantly expressed in Chinese hamster ovary (CHO) cells, and GCGR complexes were isolated by affinity purification (AP). Complexes were then analyzed by mass spectrometry (MS), and protein-GCGR interactions were validated by co-immunoprecipitation (Co-IP) and Western blot. This was followed by studies in primary hepatocytes to assess the effects of each interactor on glucagon-dependent glucose production and intracellular cAMP accumulation, and then in immortalized CHO and liver cell lines to further examine cell signaling. Thirty-three unique interactors were identified from the AP-MS screening of GCGR expressing CHO cells in both glucagon liganded and unliganded states. These studies revealed a particularly robust interaction between GCGR and 5 proteins, further validated by Co-IP, Western blot and qPCR. Overexpression of selected interactors in mouse hepatocytes indicated that two interactors, LDLR and TMED2, significantly enhanced glucagon-stimulated glucose production, while YWHAB inhibited glucose production. This was mirrored with glucagon-stimulated cAMP production, with LDLR and TMED2 enhancing and YWHAB inhibiting cAMP accumulation. To further link these interactors to glucose production, key gluconeogenic genes were assessed. Both LDLR and TMED2 stimulated while YWHAB inhibited PEPCK and G6Pase gene expression. In the present study, we have probed the GCGR interactome and found three novel GCGR interactors that control glucagon-stimulated glucose production by modulating cAMP accumulation and genes that control gluconeogenesis. These interactors may be useful targets to control glucose homeostasis in T2D.
Collapse
Affiliation(s)
- Junfeng Han
- Department of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Ming Zhang
- Department of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Sean Froese
- Department of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Feihan F. Dai
- Department of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mélanie Robitaille
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, M5S1A8, Canada
| | - Alpana Bhattacharjee
- Department of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Xinyi Huang
- Department of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Weiping Jia
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Stéphane Angers
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, M5S1A8, Canada
| | - Michael B. Wheeler
- Department of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada
- * E-mail: (MW); (LW)
| | - Li Wei
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- * E-mail: (MW); (LW)
| |
Collapse
|
17
|
Chuang CT, Guh JY, Lu CY, Wang YT, Chen HC, Chuang LY. Steap4 attenuates high glucose and S100B-induced effects in mesangial cells. J Cell Mol Med 2015; 19:1234-44. [PMID: 25817898 PMCID: PMC4459839 DOI: 10.1111/jcmm.12472] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 10/09/2014] [Indexed: 12/14/2022] Open
Abstract
Six-transmembrane epithelial antigen of prostate 4 (Steap4)-knockout mice develop hyperglycaemia and inflammation whereas Steap4 overexpression attenuates atherosclerosis in diabetic mice. Thus, we studied the roles of Steap4 in high glucose (HG, 27.5 mM) or S100B (1 μM, a ligand for the receptor for advanced glycation end-product or RAGE)-induced effects in mouse mesangial (MES13) cells. We found that HG-induced Steap4 protein expression was dependent on S100B. HG increased cell membrane, but not cytosolic, Steap4 protein expression. HG increased protein-protein interaction between Steap4 and S100B, which was confirmed by mass spectrometry of immunoprecipitated S100B. SP600125, LY294002 and AG490 attenuated S100B-induced Steap4 protein expression or gene transcriptional activity. A mutation in signal transducer and activator of transcription 3 (Stat3) site 2 of the Steap4 promoter constructs resulted in a marked decrease in HG or S100B-induced activation of Steap4 gene transcription. Overexpression of Steap4 attenuates HG or S100B-induced collagen IV, fibronectin and cyclooxygenase 2 protein expression. Overexpression of Steap4 attenuates HG or S100B-induced transforming growth factor-β (TGF-β). Moreover, overexpression of Steap4 attenuates S100B-induced signalling. Finally, overexpressing Steap4 attenuated renal expression of fibronectin, S100B, TGF-β, type IV collagen, p-Akt, p-extracellular signal regulated kinase 1/2 and p-Stat3 in streptozotocin-diabetic mice. Thus, overexpression of Steap4 attenuated HG or S100B-induced effects in MES13 cells and attenuated some of S100B-induced effects in diabetic mouse kidneys.
Collapse
Affiliation(s)
- Chao-Tang Chuang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jinn-Yuh Guh
- Department of Internal Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chi-Yu Lu
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yeng-Tseng Wang
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hung-Chun Chen
- Department of Internal Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Lea-Yea Chuang
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Center for Lipid and Glycomedicine Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
18
|
Hölper S, Nolte H, Bober E, Braun T, Krüger M. Dissection of metabolic pathways in the Db/Db mouse model by integrative proteome and acetylome analysis. MOLECULAR BIOSYSTEMS 2015; 11:908-22. [DOI: 10.1039/c4mb00490f] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
An in vivo SILAC-based quantitative proteomics approach to analyse protein abundances and acetylation levels under diabetic conditions.
Collapse
Affiliation(s)
- Soraya Hölper
- Max Planck Institute for Heart and Lung Research
- 61231 Bad Nauheim
- Germany
| | - Hendrik Nolte
- Max Planck Institute for Heart and Lung Research
- 61231 Bad Nauheim
- Germany
| | - Eva Bober
- Max Planck Institute for Heart and Lung Research
- 61231 Bad Nauheim
- Germany
| | - Thomas Braun
- Max Planck Institute for Heart and Lung Research
- 61231 Bad Nauheim
- Germany
| | - Marcus Krüger
- Max Planck Institute for Heart and Lung Research
- 61231 Bad Nauheim
- Germany
| |
Collapse
|
19
|
Karim S, Liaskou E, Fear J, Garg A, Reynolds G, Claridge L, Adams DH, Newsome PN, Lalor PF. Dysregulated hepatic expression of glucose transporters in chronic disease: contribution of semicarbazide-sensitive amine oxidase to hepatic glucose uptake. Am J Physiol Gastrointest Liver Physiol 2014; 307:G1180-90. [PMID: 25342050 PMCID: PMC4269679 DOI: 10.1152/ajpgi.00377.2013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Insulin resistance is common in patients with chronic liver disease (CLD). Serum levels of soluble vascular adhesion protein-1 (VAP-1) are also increased in these patients. The amine oxidase activity of VAP-1 stimulates glucose uptake via translocation of transporters to the cell membrane in adipocytes and smooth muscle cells. We aimed to document human hepatocellular expression of glucose transporters (GLUTs) and to determine if VAP-1 activity influences receptor expression and hepatic glucose uptake. Quantitative PCR and immunocytochemistry were used to study human liver tissue and cultured cells. We also used tissue slices from humans and VAP-1-deficient mice to assay glucose uptake and measure hepatocellular responses to stimulation. We report upregulation of GLUT1, -3, -5, -6, -7, -8, -9, -10, -11, -12, and -13 in CLD. VAP-1 expression and enzyme activity increased in disease, and provision of substrate to hepatic VAP-1 drives hepatic glucose uptake. This effect was sensitive to inhibition of VAP-1 and could be recapitulated by H2O2. VAP-1 activity also altered expression and subcellular localization of GLUT2, -4, -9, -10, and -13. Therefore, we show, for the first time, alterations in hepatocellular expression of glucose and fructose transporters in CLD and provide evidence that the semicarbazide-sensitive amine oxidase activity of VAP-1 modifies hepatic glucose homeostasis and may contribute to patterns of GLUT expression in chronic disease.
Collapse
Affiliation(s)
- Sumera Karim
- 1Centre for Liver Research and National Institute for Health Research Biomedical Research Unit, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom; and
| | - Evaggelia Liaskou
- 1Centre for Liver Research and National Institute for Health Research Biomedical Research Unit, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom; and
| | - Janine Fear
- 1Centre for Liver Research and National Institute for Health Research Biomedical Research Unit, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom; and
| | - Abhilok Garg
- 1Centre for Liver Research and National Institute for Health Research Biomedical Research Unit, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom; and
| | - Gary Reynolds
- 1Centre for Liver Research and National Institute for Health Research Biomedical Research Unit, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom; and
| | - Lee Claridge
- 1Centre for Liver Research and National Institute for Health Research Biomedical Research Unit, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom; and
| | - David H. Adams
- 1Centre for Liver Research and National Institute for Health Research Biomedical Research Unit, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom; and ,2Liver and Hepatobiliary Unit, Queen Elizabeth Hospital, Edgbaston, Birmingham, United Kingdom
| | - Philip N. Newsome
- 1Centre for Liver Research and National Institute for Health Research Biomedical Research Unit, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom; and ,2Liver and Hepatobiliary Unit, Queen Elizabeth Hospital, Edgbaston, Birmingham, United Kingdom
| | - Patricia F. Lalor
- 1Centre for Liver Research and National Institute for Health Research Biomedical Research Unit, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom; and
| |
Collapse
|
20
|
Suter MA, Ma J, Vuguin PM, Hartil K, Fiallo A, Harris RA, Charron MJ, Aagaard KM. In utero exposure to a maternal high-fat diet alters the epigenetic histone code in a murine model. Am J Obstet Gynecol 2014; 210:463.e1-463.e11. [PMID: 24793723 DOI: 10.1016/j.ajog.2014.01.045] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 01/14/2014] [Accepted: 01/31/2014] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Data from animal models show that in utero exposure to a maternal high-fat diet (HFD) renders susceptibility of these offspring to the adult onset of metabolic syndrome. We and others have previously shown that epigenetic modifications to histones may serve as a molecular memory of the in utero exposure, rendering the risk of adult disease. Because mice heterozygous for the Glut4 gene (insulin sensitive glucose transporter) born to wild-type (WT) mothers demonstrate exacterbated metabolic syndrome when exposed to an HFD in utero, we sought to analyze the genome-wide epigenetic changes that occur in the fetal liver in susceptible offspring. STUDY DESIGN WT and Glut4(+/-) (G4(+/-)) offspring of WT mothers that were exposed either to a control or an HFD in utero were studied. Immunoblotting was used to measure hepatic histone modifications of fetal and 5-week animals. Chromatin immunoprecipitation (ChIP) followed by hybridization to chip arrays (ChIP-on-chip) was used to detect genome-wide changes of histone modifications with HFD exposure. RESULTS We found that levels of hepatic H3K14ac and H3K9me3 significantly increased with HFD exposure in WT and G4(+/-) fetal and 5-week offspring. Pathway analysis of our ChIP-on-chip data revealed differential H3K14ac and H3K9me3 enrichment along pathways that regulate lipid metabolism, specifically in the promoter regions of Pparg, Ppara, Rxra, and Rora. CONCLUSION We conclude that HFD exposure in utero is associated with functional alterations to fetal hepatic histone modifications in both WT and G4(+/-) offspring, some of which persist up to 5 weeks of age.
Collapse
Affiliation(s)
- Melissa A Suter
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX
| | - Jun Ma
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX
| | - Patricia M Vuguin
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY; Division of Pediatric Endocrinology, Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY
| | - Kirsten Hartil
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY
| | - Ariana Fiallo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY
| | - R Alan Harris
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX
| | - Maureen J Charron
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY; Department of Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, NY
| | - Kjersti M Aagaard
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX.
| |
Collapse
|
21
|
Desmosterol and DHCR24: unexpected new directions for a terminal step in cholesterol synthesis. Prog Lipid Res 2013; 52:666-80. [PMID: 24095826 DOI: 10.1016/j.plipres.2013.09.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 08/25/2013] [Accepted: 09/12/2013] [Indexed: 01/31/2023]
Abstract
3β-Hydroxysterol Δ(24)-reductase (DHCR24) catalyzes the conversion of desmosterol to cholesterol. This ultimate step of cholesterol biosynthesis appears to be remarkable in its diverse functions and the number of diseases it is implicated in from vascular disease to Hepatitis C virus (HCV) infection to cancer to Alzheimer's disease. This review summarizes the present knowledge on the DHCR24 gene, sterol Δ(24)-reductase protein and the regulation of both. In addition, the functions of desmosterol, DHCR24 and their roles in human diseases are discussed. It is apparent that DHCR24 exerts more complex effects than what would be expected based on the enzymatic activity of sterol Δ(24)-reduction alone, such as its influence in modulating oxidative stress. Increasing information about DHCR24 membrane association, processing, enzymatic regulation and interaction partners will provide further fundamental insights into DHCR24 and its many and varied biological roles.
Collapse
|