1
|
Kulsange SE, Sharma M, Sonawane B, Jaiswal MR, Kulkarni MJ, Santhakumari B. SWATH-MS reveals that bisphenol A and its analogs regulate pathways leading to disruption in insulin signaling and fatty acid metabolism. Food Chem Toxicol 2024; 188:114667. [PMID: 38653447 DOI: 10.1016/j.fct.2024.114667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/24/2024] [Accepted: 04/14/2024] [Indexed: 04/25/2024]
Abstract
Bisphenol A (BPA) is an endocrine-disrupting chemical (EDC), associated with obesity and insulin resistance. The FDA prohibited the use of BPA-based polycarbonate resins in infant formula packaging; thus, its analogs, viz. Bisphenol S (BPS) and Bisphenol F (BPF) were considered alternatives in epoxy resins, plastics, and food cans. As these analogs might evoke a similar response, we investigated the role of Bisphenols (BPA, BPF, and BPS), on insulin signaling in CHO-HIRc-myc-GLUT4eGFP cells at environmentally relevant concentrations of 2 nM and 200 nM. Insulin signaling demonstrated that Bisphenols reduced phosphorylation of IR and AKT2, GLUT4 translocation, and glucose uptake. This was accompanied by increased oxidative stress. Furthermore, SWATH-MS-based proteomics of 3T3-L1 cells demonstrated that Bisphenol-treated cells regulate proteins in insulin resistance, adipogenesis, and fatty acid metabolism pathways differently. All three Bisphenols induced differentially expressed proteins enriched similar pathways, although their abundance differed for each Bisphenol. This might be due to their varying toxicity level, structural differences, and estrogen-mimetic activity. This study has important implications in addressing health concerns related to EDCs. Given that the analogs of BPA are considered alternatives to BPA, the findings of this study suggest they are equally potent in altering fatty acid metabolism and inducing insulin resistance.
Collapse
Affiliation(s)
- Shabda E Kulsange
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Monika Sharma
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Babasaheb Sonawane
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Meera R Jaiswal
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mahesh J Kulkarni
- Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - B Santhakumari
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Centre for Material Characterization, CSIR-National Chemical Laboratory, Pune 411008, India.
| |
Collapse
|
2
|
Saraswathi V, Ai W, Kumar V, Sharma K, Gopal T, Kumar N, Malhi H, Sehrawat T, Desouza CV. A Pilot Study on the Proteomics Profile of Serum Exosome-Enriched Extracellular Vesicles from Normal versus Individuals with Obesity-Related Insulin Resistance. Biomedicines 2024; 12:799. [PMID: 38672154 PMCID: PMC11048419 DOI: 10.3390/biomedicines12040799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
OBJECTIVE Circulating exosome-enriched extracellular vesicles (EVs) have drawn considerable importance in obesity-related insulin-resistance (IR). We sought to compare the proteomics profile of serum exosomes from normal individuals and those with obesity and IR. METHODS We isolated serum exosomes from male subjects with obesity and insulin resistance (Ob-IR, HOMA-IR > 2.0) and lean/overweight insulin-sensitive (Normal (N), HOMA-IR < 2.0) individuals. The differential protein expression between the two groups was detected by a label-free quantitative mass spectrometry analysis followed by GO annotation and ingenuity pathway analysis (IPA). RESULTS We identified 23 upregulated and 46 downregulated proteins between Ob-IR and N groups. Some of these proteins are involved in altering insulin signaling (VPS13C, TBC1D32, TTR, and ADIPOQ), inflammation (NFκB and CRP), and B-cell proliferation/activation (IGLV4-69, IGKV1D-13, and IGHV4-28). GO analysis revealed that the differentially expressed proteins (DEPs) are mainly involved in regulating immune cell activation and are located in extracellular space. IPA analysis showed that top molecules mediating IR, inflammation and B-cell activation were upregulated in Ob-IR subjects compared to N subjects. CONCLUSIONS Serum exosomal proteins can be used as biomarkers to identify the future risk of diabetes and a therapeutic target to prevent or slow down the progression of diabetes in high-risk individuals.
Collapse
Affiliation(s)
- Viswanathan Saraswathi
- VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (V.S.); (W.A.); (T.G.); (N.K.)
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Weilun Ai
- VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (V.S.); (W.A.); (T.G.); (N.K.)
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Vikas Kumar
- Department of Genetics Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (V.K.); (K.S.)
| | - Kanika Sharma
- Department of Genetics Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (V.K.); (K.S.)
| | - Thiyagarajan Gopal
- VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (V.S.); (W.A.); (T.G.); (N.K.)
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Narendra Kumar
- VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (V.S.); (W.A.); (T.G.); (N.K.)
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Harmeet Malhi
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA (T.S.)
| | - Tejasav Sehrawat
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA (T.S.)
| | - Cyrus V. Desouza
- VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (V.S.); (W.A.); (T.G.); (N.K.)
- Department of Internal Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
3
|
Shah R, Zhong J, Massier L, Tanriverdi K, Hwang SJ, Haessler J, Nayor M, Zhao S, Perry AS, Wilkins JT, Shadyab AH, Manson JE, Martin L, Levy D, Kooperberg C, Freedman JE, Rydén M, Murthy VL. Targeted Proteomics Reveals Functional Targets for Early Diabetes Susceptibility in Young Adults. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2024; 17:e004192. [PMID: 38323454 PMCID: PMC10940209 DOI: 10.1161/circgen.123.004192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 11/05/2023] [Indexed: 02/08/2024]
Abstract
BACKGROUND The circulating proteome may encode early pathways of diabetes susceptibility in young adults for surveillance and intervention. Here, we define proteomic correlates of tissue phenotypes and diabetes in young adults. METHODS We used penalized models and principal components analysis to generate parsimonious proteomic signatures of diabetes susceptibility based on phenotypes and on diabetes diagnosis across 184 proteins in >2000 young adults in the CARDIA (Coronary Artery Risk Development in Young Adults study; mean age, 32 years; 44% women; 43% Black; mean body mass index, 25.6±4.9 kg/m2), with validation against diabetes in >1800 individuals in the FHS (Framingham Heart Study) and WHI (Women's Health Initiative). RESULTS In 184 proteins in >2000 young adults in CARDIA, we identified 2 proteotypes of diabetes susceptibility-a proinflammatory fat proteotype (visceral fat, liver fat, inflammatory biomarkers) and a muscularity proteotype (muscle mass), linked to diabetes in CARDIA and WHI/FHS. These proteotypes specified broad mechanisms of early diabetes pathogenesis, including transorgan communication, hepatic and skeletal muscle stress responses, vascular inflammation and hemostasis, fibrosis, and renal injury. Using human adipose tissue single cell/nuclear RNA-seq, we demonstrate expression at transcriptional level for implicated proteins across adipocytes and nonadipocyte cell types (eg, fibroadipogenic precursors, immune and vascular cells). Using functional assays in human adipose tissue, we demonstrate the association of expression of genes encoding these implicated proteins with adipose tissue metabolism, inflammation, and insulin resistance. CONCLUSIONS A multifaceted discovery effort uniting proteomics, underlying clinical susceptibility phenotypes, and tissue expression patterns may uncover potentially novel functional biomarkers of early diabetes susceptibility in young adults for future mechanistic evaluation.
Collapse
Affiliation(s)
- Ravi Shah
- Vanderbilt Translational & Clinical Cardiovascular Research Center, Vanderbilt Univ, Nashville, TN
| | - Jiawei Zhong
- Dept of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | - Lucas Massier
- Dept of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | - Kahraman Tanriverdi
- Vanderbilt Translational & Clinical Cardiovascular Research Center, Vanderbilt Univ, Nashville, TN
| | - Shih-Jen Hwang
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | - Matthew Nayor
- Sections of Preventive Medicine & Epidemiology & Cardiovascular Medicine, Dept of Medicine, Dept of Epidemiology, Boston University Schools of Medicine & Public Health, Boston, MA & Framingham Heart Study, Framingham, MA
| | | | - Andrew S. Perry
- Vanderbilt Translational & Clinical Cardiovascular Research Center, Vanderbilt Univ, Nashville, TN
| | | | - Aladdin H. Shadyab
- Herbert Wertheim School of Public Health & Human Longevity Science, Univ of California, San Diego, La Jolla, CA
| | - JoAnn E. Manson
- Dept of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Lisa Martin
- George Washington Univ School of Medicine & Health Sciences
| | - Daniel Levy
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | - Jane E. Freedman
- Vanderbilt Translational & Clinical Cardiovascular Research Center, Vanderbilt Univ, Nashville, TN
| | - Mikael Rydén
- Dept of Medicine (H7), Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
4
|
Larsen JK, Kruse R, Sahebekhtiari N, Moreno-Justicia R, Gomez Jorba G, Petersen MH, de Almeida ME, Ørtenblad N, Deshmukh AS, Højlund K. High-throughput proteomics uncovers exercise training and type 2 diabetes-induced changes in human white adipose tissue. SCIENCE ADVANCES 2023; 9:eadi7548. [PMID: 38019916 PMCID: PMC10686561 DOI: 10.1126/sciadv.adi7548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023]
Abstract
White adipose tissue (WAT) is important for metabolic homeostasis. We established the differential proteomic signatures of WAT in glucose-tolerant lean and obese individuals and patients with type 2 diabetes (T2D) and the response to 8 weeks of high-intensity interval training (HIIT). Using a high-throughput and reproducible mass spectrometry-based proteomics pipeline, we identified 3773 proteins and found that most regulated proteins displayed progression in markers of dysfunctional WAT from lean to obese to T2D individuals and were highly associated with clinical measures such as insulin sensitivity and HbA1c. We propose that these distinct markers could serve as potential clinical biomarkers. HIIT induced only minor changes in the WAT proteome. This included an increase in WAT ferritin levels independent of obesity and T2D, and WAT ferritin levels were strongly correlated with individual insulin sensitivity. Together, we report a proteomic signature of WAT related to obesity and T2D and highlight an unrecognized role of human WAT iron metabolism in exercise training adaptations.
Collapse
Affiliation(s)
- Jeppe Kjærgaard Larsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Rikke Kruse
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Navid Sahebekhtiari
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| | - Roger Moreno-Justicia
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Gerard Gomez Jorba
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Maria H. Petersen
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, Denmark
| | - Martin E. de Almeida
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, Denmark
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Niels Ørtenblad
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Atul S. Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense C, Denmark
| |
Collapse
|
5
|
Sabaratnam R, Hansen DR, Svenningsen P. White adipose tissue mitochondrial bioenergetics in metabolic diseases. Rev Endocr Metab Disord 2023; 24:1121-1133. [PMID: 37558853 DOI: 10.1007/s11154-023-09827-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/19/2023] [Indexed: 08/11/2023]
Abstract
White adipose tissue (WAT) is an important endocrine organ that regulates systemic energy metabolism. In metabolically unhealthy obesity, adipocytes become dysfunctional through hypertrophic mechanisms associated with a reduced endocrine function, reduced mitochondrial function, but increased inflammation, fibrosis, and extracellular remodelling. A pathologic WAT remodelling promotes systemic lipotoxicity characterized by fat accumulation in tissues such as muscle and liver, leading to systemic insulin resistance and type 2 diabetes. Several lines of evidence from human and animal studies suggest a link between unhealthy obesity and adipocyte mitochondrial dysfunction, and interventions that improve mitochondrial function may reduce the risk of obesity-associated diseases. This review discusses the importance of mitochondrial function and metabolism in human adipocyte biology and intercellular communication mechanisms within WAT. Moreover, a selected interventional approach for better adipocyte mitochondrial metabolism in humans is reviewed. A greater understanding of mitochondrial bioenergetics in WAT might provide novel therapeutic opportunities to prevent or restore dysfunctional adipose tissue in obesity-associated diseases.
Collapse
Affiliation(s)
- Rugivan Sabaratnam
- Department of Clinical Research, University of Southern Denmark, Odense C, DK-5000, Denmark.
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, DK-5000, Denmark.
- Department of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, J. B. Winsløws Vej 21,3, Odense C, DK-5000, Denmark.
| | - Didde Riisager Hansen
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, DK-5000, Denmark
- Department of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, J. B. Winsløws Vej 21,3, Odense C, DK-5000, Denmark
| | - Per Svenningsen
- Department of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, J. B. Winsløws Vej 21,3, Odense C, DK-5000, Denmark.
| |
Collapse
|
6
|
Liu X, Zheng T, Tao MY, Huang R, Zhang GH, Yang MN, Xu YJ, Wang WJ, He H, Fang F, Dong Y, Fan JG, Zhang J, Ouyang F, Li F, Luo ZC. Cord blood fatty acid binding protein 4 and lipids in infants born small- or large-for-gestational-age. Front Pediatr 2023; 11:1078048. [PMID: 37274820 PMCID: PMC10237290 DOI: 10.3389/fped.2023.1078048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 04/28/2023] [Indexed: 06/07/2023] Open
Abstract
Aim Adverse (poor or excessive) fetal growth "programs" an elevated risk of type 2 diabetes. Fatty acid binding protein 4 (FABP4) has been implicated in regulating insulin sensitivity and lipid metabolism relevant to fetal growth. We sought to determine whether FABP4 is associated with poor or excessive fetal growth and fetal lipids. Methods In a nested case-control study in the Shanghai Birth Cohort including 60 trios of small-for-gestational-age (SGA, an indicator of poor fetal growth), large-for-gestational-age (LGA, an indicator of excessive fetal growth) and optimal-for-gestational-age (OGA, control) infants, we measured cord blood concentrations of FABP4 and lipids [high-density lipoprotein (HDL) and low-density lipoprotein (LDL) cholesterols, triglycerides (TG)]. Results Adjusting for maternal and neonatal characteristics, higher cord blood FABP4 concentrations were associated with a lower odds of SGA [OR = 0.29 (0.11-0.77) per log unit increment in FABP4, P = 0.01], but were not associated with LGA (P = 0.46). Cord blood FABP4 was positively correlated with both LDL (r = 0.29, P = 0.025) and HDL (r = 0.33, P = 0.01) in LGA infants only. Conclusion FABP4 was inversely associated with the risk of SGA. The study is the first to demonstrate LGA-specific positive correlations of cord blood FABP4 with HDL and LDL cholesterols, suggesting a role of FABP4 in fetal lipid metabolism in subjects with excessive fetal growth.
Collapse
Affiliation(s)
- Xin Liu
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
- Lunenfeld-Tanenbaum Research Institute, Prosserman Centre for Population Health Research, Department of Obstetrics and Gynecology, Mount Sinai Hospital, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Tao Zheng
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Min-Yi Tao
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Rong Huang
- Lunenfeld-Tanenbaum Research Institute, Prosserman Centre for Population Health Research, Department of Obstetrics and Gynecology, Mount Sinai Hospital, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Guang-Hui Zhang
- Department of Clinical Assay Laboratory, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Meng-Nan Yang
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Ya-Jie Xu
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Wen-Juan Wang
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
- Clinical Skills Center, School of Clinical Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Hua He
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Fang Fang
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Yu Dong
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Jian-Gao Fan
- Center for Fatty Liver, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Zhang
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Fengxiu Ouyang
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Fei Li
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Zhong-Cheng Luo
- Ministry of Education-Shanghai Key Laboratory of Children’s Environmental Health, Early Life Health Institute, and Department of Pediatrics, Xinhua Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
- Lunenfeld-Tanenbaum Research Institute, Prosserman Centre for Population Health Research, Department of Obstetrics and Gynecology, Mount Sinai Hospital, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
7
|
Youxiang C, Lin Z, Zekai C, Weijun X. Resting and exercise metabolic characteristics in obese children with insulin resistance. Front Physiol 2022; 13:1049560. [DOI: 10.3389/fphys.2022.1049560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022] Open
Abstract
Purpose: This study aimed to explore the characteristics of resting energy expenditure (REE) and lipid metabolism during incremental load exercise in obese children and adolescents with insulin resistance (IR) to provide evidence for exercise intervention in obese children and adolescents with IR.Method: From July 2019 to August 2021, 195 obese children and adolescents aged 13–17 were recruited through a summer camp. The participants were divided into IR (n = 67) and no-IR (without insulin resistance, n = 128) groups and underwent morphology, blood indicators, body composition, and resting energy consumption gas metabolism tests. Thirty participants each were randomly selected from the IR and no-IR groups to carry out the incremental treadmill test.Results: Significant metabolic differences in resting and exercise duration were found between the IR and no-IR groups. In the resting state, the resting metabolic equivalents (4.33 ± 0.94 ml/min/kg vs. 3.91 ± 0.73 ml/min/kg, p = 0.001) and REE (2464.03 ± 462.29 kcal/d vs. 2143.88 ± 380.07 kcal/d, p < 0.001) in the IR group were significantly higher than in the no-IR group. During exercise, the absolute maximal fat oxidation (0.33 ± 0.07 g/min vs. 0.36 ± 0.09 g/min, p = 0.002) in the IR group was significantly lower than in the no-IR group; maximal fat oxidation intensity (130.9 ± 8.9 bpm vs. 139.9 ± 7.4 bpm, p = 0.040) was significantly lower in the IR group.Conclusion: Significant resting and exercise metabolic differences were found between obese IR and no-IR children and adolescents. Obese IR children and adolescents have higher REE and lower maximal fat oxidation intensity than obese no-IR children and adolescents.
Collapse
|
8
|
Lai W, Shi M, Huang R, Fu P, Ma L. Fatty acid-binding protein 4 in kidney diseases: From mechanisms to clinics. Eur J Pharmacol 2022; 931:175224. [PMID: 35995212 DOI: 10.1016/j.ejphar.2022.175224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/28/2022] [Accepted: 08/16/2022] [Indexed: 11/03/2022]
Abstract
Considerable evidence indicated the relationship between fatty acid-binding protein 4 (FABP4) and kidney diseases. FABP4, a small molecular lipid chaperone, is identified to regulate fatty acid oxidation, inflammation, apoptosis, endoplasmic reticulum stress and macrophage-to-myofibroblast transition in kidney diseases. Many studies have shown that circulating FABP4 level is related to proteinuria, renal function decline, cardiovascular complications of end-stage renal disease and even the prognosis of kidney transplanted patients. Notably, pharmacological or genetic inhibition of FABP4 attenuated renal injury in the various experimental models of kidney diseases, making it promising to develop potential therapeutic strategies targeting FABP4 in kidney diseases. In this study, we updated and reviewed the mechanisms and clinical significance of FABP4 in kidney diseases.
Collapse
Affiliation(s)
- Weijing Lai
- Kidney Research Institute, Department of Nephrology, West China Hospital of Sichuan University, Sichuan, Chengdu, 610041, China; Department of Nephrology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Min Shi
- Kidney Research Institute, Department of Nephrology, West China Hospital of Sichuan University, Sichuan, Chengdu, 610041, China
| | - Rongshuang Huang
- Kidney Research Institute, Department of Nephrology, West China Hospital of Sichuan University, Sichuan, Chengdu, 610041, China
| | - Ping Fu
- Kidney Research Institute, Department of Nephrology, West China Hospital of Sichuan University, Sichuan, Chengdu, 610041, China.
| | - Liang Ma
- Kidney Research Institute, Department of Nephrology, West China Hospital of Sichuan University, Sichuan, Chengdu, 610041, China.
| |
Collapse
|
9
|
Sabaratnam R, Skov V, Paulsen SK, Juhl S, Kruse R, Hansen T, Halkier C, Kristensen JM, Vind BF, Richelsen B, Knudsen S, Dahlgaard J, Beck-Nielsen H, Kruse TA, Højlund K. A Signature of Exaggerated Adipose Tissue Dysfunction in Type 2 Diabetes Is Linked to Low Plasma Adiponectin and Increased Transcriptional Activation of Proteasomal Degradation in Muscle. Cells 2022; 11:cells11132005. [PMID: 35805088 PMCID: PMC9265693 DOI: 10.3390/cells11132005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/12/2022] [Accepted: 06/21/2022] [Indexed: 01/27/2023] Open
Abstract
Insulin resistance in skeletal muscle in type 2 diabetes (T2D) is characterized by more pronounced metabolic and molecular defects than in obesity per se. There is increasing evidence that adipose tissue dysfunction contributes to obesity-induced insulin resistance in skeletal muscle. Here, we used an unbiased approach to examine if adipose tissue dysfunction is exaggerated in T2D and linked to diabetes-related mechanisms of insulin resistance in skeletal muscle. Transcriptional profiling and biological pathways analysis were performed in subcutaneous adipose tissue (SAT) and skeletal muscle biopsies from 17 patients with T2D and 19 glucose-tolerant, age and weight-matched obese controls. Findings were validated by qRT-PCR and western blotting of selected genes and proteins. Patients with T2D were more insulin resistant and had lower plasma adiponectin than obese controls. Transcriptional profiling showed downregulation of genes involved in mitochondrial oxidative phosphorylation and the tricarboxylic-acid cycle and increased expression of extracellular matrix (ECM) genes in SAT in T2D, whereas genes involved in proteasomal degradation were upregulated in the skeletal muscle in T2D. qRT-PCR confirmed most of these findings and showed lower expression of adiponectin in SAT and higher expression of myostatin in muscle in T2D. Interestingly, muscle expression of proteasomal genes correlated positively with SAT expression of ECM genes but inversely with the expression of ADIPOQ in SAT and plasma adiponectin. Protein content of proteasomal subunits and major ubiquitin ligases were unaltered in the skeletal muscle of patients with T2D. A transcriptional signature of exaggerated adipose tissue dysfunction in T2D, compared with obesity alone, is linked to low plasma adiponectin and increased transcriptional activation of proteasomal degradation in skeletal muscle.
Collapse
Affiliation(s)
- Rugivan Sabaratnam
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; (T.H.); (C.H.)
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, DK-4000 Roskilde, Denmark;
| | - Søren K. Paulsen
- Department of Pathology, Viborg Regional Hospital, DK-8800 Viborg, Denmark;
| | - Stine Juhl
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; (T.H.); (C.H.)
| | - Rikke Kruse
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; (T.H.); (C.H.)
| | - Thea Hansen
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; (T.H.); (C.H.)
| | - Cecilie Halkier
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; (T.H.); (C.H.)
| | - Jonas M. Kristensen
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
- Molecular Physiology Section, Department of Nutrition, Exercise and Sports, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Birgitte F. Vind
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
| | - Bjørn Richelsen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, DK-8200 Aarhus N, Denmark;
| | - Steen Knudsen
- Allarity Therapeutics Europe, DK-2970 Hørsholm, Denmark;
| | - Jesper Dahlgaard
- Program for Mind and Body in Mental Health, Research Centre for Health and Welfare Technology, VIA University College, DK-8200 Aarhus, Denmark;
- Department of Clinical Medicine, Aarhus University, DK-8200 Aarhus, Denmark
| | - Henning Beck-Nielsen
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
| | - Torben A. Kruse
- Department of Clinical Genetics, Odense University Hospital, DK-5000 Odense C, Denmark;
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; (T.H.); (C.H.)
- Correspondence: ; Tel.: +45-2532-0648
| |
Collapse
|
10
|
Ghozlan H, Cox A, Nierenberg D, King S, Khaled AR. The TRiCky Business of Protein Folding in Health and Disease. Front Cell Dev Biol 2022; 10:906530. [PMID: 35602608 PMCID: PMC9117761 DOI: 10.3389/fcell.2022.906530] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/20/2022] [Indexed: 01/03/2023] Open
Abstract
Maintenance of the cellular proteome or proteostasis is an essential process that when deregulated leads to diseases like neurological disorders and cancer. Central to proteostasis are the molecular chaperones that fold proteins into functional 3-dimensional (3D) shapes and prevent protein aggregation. Chaperonins, a family of chaperones found in all lineages of organisms, are efficient machines that fold proteins within central cavities. The eukaryotic Chaperonin Containing TCP1 (CCT), also known as Tailless complex polypeptide 1 (TCP-1) Ring Complex (TRiC), is a multi-subunit molecular complex that folds the obligate substrates, actin, and tubulin. But more than folding cytoskeletal proteins, CCT differs from most chaperones in its ability to fold proteins larger than its central folding chamber and in a sequential manner that enables it to tackle proteins with complex topologies or very large proteins and complexes. Unique features of CCT include an asymmetry of charges and ATP affinities across the eight subunits that form the hetero-oligomeric complex. Variable substrate binding capacities endow CCT with a plasticity that developed as the chaperonin evolved with eukaryotes and acquired functional capacity in the densely packed intracellular environment. Given the decades of discovery on the structure and function of CCT, much remains unknown such as the scope of its interactome. New findings on the role of CCT in disease, and potential for diagnostic and therapeutic uses, heighten the need to better understand the function of this essential molecular chaperone. Clues as to how CCT causes cancer or neurological disorders lie in the early studies of the chaperonin that form a foundational knowledgebase. In this review, we span the decades of CCT discoveries to provide critical context to the continued research on the diverse capacities in health and disease of this essential protein-folding complex.
Collapse
Affiliation(s)
- Heba Ghozlan
- Division of Cancer Research, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
- Department of Physiology and Biochemistry, Jordan University of Science and Technology, Irbid, Jordan
| | - Amanda Cox
- Division of Cancer Research, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Daniel Nierenberg
- Division of Cancer Research, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Stephen King
- Division of Neuroscience, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Annette R. Khaled
- Division of Cancer Research, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| |
Collapse
|
11
|
Georgiev A, Granata C, Roden M. The role of mitochondria in the pathophysiology and treatment of common metabolic diseases in humans. Am J Physiol Cell Physiol 2022; 322:C1248-C1259. [PMID: 35508191 DOI: 10.1152/ajpcell.00035.2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Common metabolic diseases such as obesity, type 2 diabetes mellitus and non-alcoholic fatty liver disease significantly contribute to morbidity and mortality worldwide. They frequently associate with insulin resistance and altered mitochondrial functionality. Insulin-responsive tissues can show changes in mitochondrial features such as oxidative capacity, mitochondrial content and turnover, which do not necessarily reflect abnormalities but rather adaption to a certain metabolic condition. Lifestyle modifications and classic or novel drugs can modify these alterations and help treating these metabolic diseases. This review addresses the role of mitochondria in human metabolic diseases and discusses potential future research directions.
Collapse
Affiliation(s)
- Asen Georgiev
- Institute for Clinical Diabetology, German, Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Cesare Granata
- Institute for Clinical Diabetology, German, Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany.,Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Institute for Health and Sport (iHeS), Victoria University, Melbourne, VIC, Australia
| | - Michael Roden
- Institute for Clinical Diabetology, German, Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany.,Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
12
|
Localization of aquaglyceroporins in human and murine white adipose tissue. Histochem Cell Biol 2022; 157:623-639. [PMID: 35235046 DOI: 10.1007/s00418-022-02090-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2022] [Indexed: 11/04/2022]
Abstract
The glycerol channel AQP7 facilitates glycerol efflux from adipose tissue (AT), and AQP7 deficiency has been suggested to promote obesity. However, the release of glycerol from AT is not fully blocked in AQP7-deficient mice, which suggests that either alternative glycerol channels are present in AT or significant simple diffusion of glycerol occurs. Previous investigations of the expression of other aquaglyceroporins (AQP3, AQP9, AQP10) than AQP7 in AT are contradictory. Therefore, we here aim at determining the cellular localization of AQP3 and AQP9 in addition to AQP7 in human and mouse AT using well-characterized antibodies for immunohistochemistry (IHC) and immunoblotting as well as available single-cell transcriptomic data from human and mouse AT. We confirm that AQP7 is expressed in endothelial cells and adipocytes in human AT and find ex vivo evidence for interaction between AQP7 and perilipin-1 in adipocytes. In addition, labeling for AQP7 in human AT also includes CD68-positive cells. No labeling for AQP3 or AQP9 was identified in endothelial cells or adipocytes in human or mouse AT using IHC. Instead, in human AT, AQP3 was predominantly found in erythrocytes, whereas AQP9 expression was observed in a small number of CD15-positive cells. The transcriptomic data revealed that AQP3 mRNA was found in a low number of cells in most of the identified cell clusters, whereas AQP9 mRNA was found in myeloid cell clusters as well as in clusters likely representing mesothelial progenitor cells. No AQP10 mRNA was identified in human AT. In conclusion, the presented results do not suggest a functional overlap between AQP3/AQP9/AQP10 and AQP7 in human or mouse white AT.
Collapse
|
13
|
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
|
14
|
Van Meijel RLJ, Wang P, Bouwman F, Blaak EE, Mariman ECM, Goossens GH. The Effects of Mild Intermittent Hypoxia Exposure on the Abdominal Subcutaneous Adipose Tissue Proteome in Overweight and Obese Men: A First-in-Human Randomized, Single-Blind, and Cross-Over Study. Front Physiol 2022; 12:791588. [PMID: 35058800 PMCID: PMC8764283 DOI: 10.3389/fphys.2021.791588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Adipose tissue (AT) oxygen tension (pO2) has been implicated in AT dysfunction and metabolic perturbations in both rodents and humans. Compelling evidence suggests that hypoxia exposure alters metabolism, at least partly through effects on AT. However, it remains to be elucidated whether mild intermittent hypoxia (MIH) exposure impacts the AT proteome. We performed a randomized, single-blind, and cross-over study to investigate the effects of seven consecutive days of MIH (FiO2 15%, 3x2h/d) compared to normoxia (FiO2 21%) exposure on the AT proteome in overweight/obese men. In vivo AT insulin sensitivity was determined by the gold standard hyperinsulinemic-euglycemic clamp, and abdominal subcutaneous AT biopsies were collected under normoxic fasting conditions following both exposure regimens (day 8). AT proteins were isolated and quantified using liquid chromatography-mass spectrometry. After correction for blood contamination, 1,022 AT protein IDs were identified, of which 123 were differentially expressed following MIH (p < 0.05). We demonstrate for the first time that MIH exposure, which markedly reduces in vivo AT oxygen tension, impacts the human AT proteome. Although we cannot exclude that a single differentially expressed protein might be a false positive finding, several functional pathways were altered by MIH exposure, also after adjustment for multiple testing. Specifically, differentially expressed proteins were involved in redox systems, cell-adhesion, actin cytoskeleton organization, extracellular matrix composition, and energy metabolism. The MIH-induced change in AT TMOD3 expression was strongly related to altered in vivo AT insulin sensitivity, thus linking MIH-induced effects on the AT proteome to metabolic changes in overweight/obese humans.
Collapse
Affiliation(s)
- Rens L J Van Meijel
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Ping Wang
- Department of Clinical Genetics, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Freek Bouwman
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Edwin C M Mariman
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| |
Collapse
|
15
|
Flenkenthaler F, Ländström E, Shashikadze B, Backman M, Blutke A, Philippou-Massier J, Renner S, Hrabe de Angelis M, Wanke R, Blum H, Arnold GJ, Wolf E, Fröhlich T. Differential Effects of Insulin-Deficient Diabetes Mellitus on Visceral vs. Subcutaneous Adipose Tissue-Multi-omics Insights From the Munich MIDY Pig Model. Front Med (Lausanne) 2021; 8:751277. [PMID: 34888323 PMCID: PMC8650062 DOI: 10.3389/fmed.2021.751277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 10/25/2021] [Indexed: 12/18/2022] Open
Abstract
Adipose tissue (AT) is no longer considered to be responsible for energy storage only but is now recognized as a major endocrine organ that is distributed across different parts of the body and is actively involved in regulatory processes controlling energy homeostasis. Moreover, AT plays a crucial role in the development of metabolic disease such as diabetes. Recent evidence has shown that adipokines have the ability to regulate blood glucose levels and improve metabolic homeostasis. While AT has been studied extensively in the context of type 2 diabetes, less is known about how different AT types are affected by absolute insulin deficiency in type 1 or permanent neonatal diabetes mellitus. Here, we analyzed visceral and subcutaneous AT in a diabetic, insulin-deficient pig model (MIDY) and wild-type (WT) littermate controls by RNA sequencing and quantitative proteomics. Multi-omics analysis indicates a depot-specific dysregulation of crucial metabolic pathways in MIDY AT samples. We identified key proteins involved in glucose uptake and downstream signaling, lipogenesis, lipolysis and β-oxidation to be differentially regulated between visceral and subcutaneous AT in response to insulin deficiency. Proteins related to glycogenolysis, pyruvate metabolism, TCA cycle and lipogenesis were increased in subcutaneous AT, whereas β-oxidation-related proteins were increased in visceral AT from MIDY pigs, pointing at a regionally different metabolic adaptation to master energy stress arising from diminished glucose utilization in MIDY AT. Chronic, absolute insulin deficiency and hyperglycemia revealed fat depot-specific signatures using multi-omics analysis. The generated datasets are a valuable resource for further comparative and translational studies in clinical diabetes research.
Collapse
Affiliation(s)
- Florian Flenkenthaler
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany.,German Center for Diabetes Research (DZD), Oberschleißheim, Germany
| | - Erik Ländström
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany.,Gene Center, Graduate School of Quantitative Biosciences Munich (QBM), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Bachuki Shashikadze
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Mattias Backman
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany.,Gene Center, Graduate School of Quantitative Biosciences Munich (QBM), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Andreas Blutke
- Helmholtz Zentrum München, Institute of Experimental Genetics, Oberschleißheim, Germany
| | - Julia Philippou-Massier
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany.,German Center for Diabetes Research (DZD), Oberschleißheim, Germany
| | - Simone Renner
- German Center for Diabetes Research (DZD), Oberschleißheim, Germany.,Department of Veterinary Sciences, Gene Center, Institute for Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), Ludwig-Maximilians-Universität (LMU) Munich, Oberschleißheim, Germany
| | - Martin Hrabe de Angelis
- German Center for Diabetes Research (DZD), Oberschleißheim, Germany.,Helmholtz Zentrum München, Institute of Experimental Genetics, Technical University of Munich, Munich, Germany
| | - Rüdiger Wanke
- Center for Clinical Veterinary Medicine, Institute of Veterinary Pathology, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Georg J Arnold
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Eckhard Wolf
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany.,German Center for Diabetes Research (DZD), Oberschleißheim, Germany.,Department of Veterinary Sciences, Gene Center, Institute for Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), Ludwig-Maximilians-Universität (LMU) Munich, Oberschleißheim, Germany
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| |
Collapse
|
16
|
Abstract
Lipid droplets (LDs) are endoplasmic reticulum-derived organelles that consist of a core of neutral lipids encircled by a phospholipid monolayer decorated with proteins. As hubs of cellular lipid and energy metabolism, LDs are inherently involved in the etiology of prevalent metabolic diseases such as obesity and nonalcoholic fatty liver disease. The functions of LDs are regulated by a unique set of associated proteins, the LD proteome, which includes integral membrane and peripheral proteins. These proteins control key activities of LDs such as triacylglycerol synthesis and breakdown, nutrient sensing and signal integration, and interactions with other organelles. Here we review the mechanisms that regulate the composition of the LD proteome, such as pathways that mediate selective and bulk LD protein degradation and potential connections between LDs and cellular protein quality control.
Collapse
Affiliation(s)
- Melissa A Roberts
- Department of Molecular and Cell Biology and Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720, USA;
| | - James A Olzmann
- Department of Molecular and Cell Biology and Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720, USA; .,Chan Zuckerberg Biohub, San Francisco, California 94158, USA
| |
Collapse
|
17
|
Raajendiran A, Krisp C, Souza DPD, Ooi G, Burton PR, Taylor RA, Molloy MP, Watt MJ. Proteome analysis of human adipocytes identifies depot-specific heterogeneity at metabolic control points. Am J Physiol Endocrinol Metab 2021; 320:E1068-E1084. [PMID: 33843278 DOI: 10.1152/ajpendo.00473.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Adipose tissue is a primary regulator of energy balance and metabolism. The distribution of adipose tissue depots is of clinical interest because the accumulation of upper-body subcutaneous (ASAT) and visceral adipose tissue (VAT) is associated with cardiometabolic diseases, whereas lower-body glutealfemoral adipose tissue (GFAT) appears to be protective. There is heterogeneity in morphology and metabolism of adipocytes obtained from different regions of the body, but detailed knowledge of the constituent proteins in each depot is lacking. Here, we determined the human adipocyte proteome from ASAT, VAT, and GFAT using high-resolution Sequential Window Acquisition of all Theoretical (SWATH) mass spectrometry proteomics. We quantified 4,220 proteins in adipocytes, and 2,329 proteins were expressed in all three adipose depots. Comparative analysis revealed significant differences between adipocytes from different regions (6% and 8% when comparing VAT vs. ASAT and GFAT, 3% when comparing the subcutaneous adipose tissue depots, ASAT and GFAT), with marked differences in proteins that regulate metabolic functions. The VAT adipocyte proteome was overrepresented with proteins of glycolysis, lipogenesis, oxidative stress, and mitochondrial dysfunction. The GFAT adipocyte proteome predicted the activation of peroxisome proliferator-activated receptor α (PPARα), fatty acid, and branched-chain amino acid (BCAA) oxidation, enhanced tricarboxylic acid (TCA) cycle flux, and oxidative phosphorylation, which was supported by metabolomic data obtained from adipocytes. Together, this proteomic analysis provides an important resource and novel insights that enhance the understanding of metabolic heterogeneity in the regional adipocytes of humans.NEW & NOTEWORTHY Adipocyte metabolism varies depending on anatomical location and the adipocyte protein composition may orchestrate this heterogeneity. We used SWATH proteomics in patient-matched human upper- (visceral and subcutaneous) and lower-body (glutealfemoral) adipocytes and detected 4,220 proteins and distinguishable regional proteomes. Upper-body adipocyte proteins were associated with glycolysis, de novo lipogenesis, mitochondrial dysfunction, and oxidative stress, whereas lower-body adipocyte proteins were associated with enhanced PPARα activation, fatty acid, and BCAA oxidation, TCA cycle flux, and oxidative phosphorylation.
Collapse
Affiliation(s)
- Arthe Raajendiran
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
- Department of Physiology, Monash University, Clayton, Victoria, Australia
- Metabolism, Diabetes and Obesity Program, Monash Biomedicine Discovery Institute, University of Melbourne, Melbourne, Victoria, Australia
| | - Christoph Krisp
- Australian Proteome Analysis Facility, Macquarie University, New South Wales, Australia
| | - David P De Souza
- Metabolomics Australia, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Parkville, Victoria, Australia
| | - Geraldine Ooi
- Faculty of Medicine, Nursing and Health Sciences, Centre for Obesity Research and Education, Monash University, Melbourne, Victoria, Australia
| | - Paul R Burton
- Faculty of Medicine, Nursing and Health Sciences, Centre for Obesity Research and Education, Monash University, Melbourne, Victoria, Australia
| | - Renea A Taylor
- Department of Physiology, Monash University, Clayton, Victoria, Australia
- Cancer Research Division, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - Mark P Molloy
- Australian Proteome Analysis Facility, Macquarie University, New South Wales, Australia
| | - Matthew J Watt
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
18
|
Jin C, Lin L, Han N, Zhao Z, Xu X, Luo S, Liu J, Wang H. Risk of Gestational Diabetes Mellitus in relation to Plasma Concentrations of Fatty Acid-Binding Protein 4: A Nested Case-Control Study in China. J Diabetes Res 2021; 2021:6681432. [PMID: 34368366 PMCID: PMC8342154 DOI: 10.1155/2021/6681432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 07/08/2021] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE The study is aimed at examining the effects of fatty acid-binding protein 4 (FABP4) on insulin resistance and gestational diabetes mellitus (GDM). METHODS Based on a prospective birth cohort in Beijing, China, we conducted a nested case-control study and analyzed 135 GDM case-control pairs matched by age and the gestational week when they took the oral glucose tolerance test. We performed linear regression to analyze the association of plasma FABP4 concentrations with insulin resistance. We used logistic regression to estimate odds ratios (ORs) of FABP4 for GDM, controlling for potential confounders, including dietary intake and physical activity. RESULTS Plasma FABP4 levels in the first and second trimesters were positively associated with fasting insulin and homeostasis model assessment for insulin resistance (HOMA-IR) in the second trimester (both P < 0.001). Compared with those in the lowest FABP4 tertile, women in the highest tertile of FABP4 levels in the first and second trimesters had 1.053 times (OR = 2.053, 95% CI 1.091 to 3.863) and 1.447 times (OR = 2.447, 95% CI 1.305 to 4.588) higher risk of developing GDM. CONCLUSIONS Elevated FABP4 levels in the first and second trimesters were associated with a higher level of insulin resistance and greater GDM risk, indicating FABP4 might predict women with high risk of developing GDM.
Collapse
Affiliation(s)
- Chuyao Jin
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing, China
| | - Lizi Lin
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing, China
| | - Na Han
- Tongzhou Maternal and Child Health Hospital, Beijing, China
| | - Zhiling Zhao
- Tongzhou Maternal and Child Health Hospital, Beijing, China
| | - Xiangrong Xu
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing, China
| | - Shusheng Luo
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing, China
| | - Jue Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Haijun Wang
- Department of Maternal and Child Health, School of Public Health, Peking University, Beijing, China
| |
Collapse
|
19
|
Bódis K, Jelenik T, Lundbom J, Markgraf DF, Strom A, Zaharia OP, Karusheva Y, Burkart V, Müssig K, Kupriyanova Y, Ouni M, Wolkersdorfer M, Hwang JH, Ziegler D, Schürmann A, Roden M, Szendroedi J. Expansion and Impaired Mitochondrial Efficiency of Deep Subcutaneous Adipose Tissue in Recent-Onset Type 2 Diabetes. J Clin Endocrinol Metab 2020; 105:5678088. [PMID: 31838512 PMCID: PMC7060761 DOI: 10.1210/clinem/dgz267] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 12/13/2019] [Indexed: 01/21/2023]
Abstract
CONTEXT/OBJECTIVE Impaired adipose tissue (AT) function might induce recent-onset type 2 diabetes (T2D). Understanding AT energy metabolism could yield novel targets for the treatment of T2D. DESIGN/PATIENTS Male patients with recently-diagnosed T2D and healthy male controls (CON) of similar abdominal subcutaneous AT (SAT)-thickness, fat mass, and age (n = 14 each), underwent hyperinsulinemic-euglycemic clamps with [6,6-2H2]glucose and indirect calorimetry. We assessed mitochondrial efficiency (coupling: state 3/4o; proton leak: state 4o/u) via high-resolution respirometry in superficial (SSAT) and deep (DSAT) SAT-biopsies, hepatocellular lipids (HCL) and fat mass by proton-magnetic-resonance-spectroscopy and -imaging. RESULTS T2D patients (known diabetes duration: 2.5 [0.1; 5.0] years) had 43%, 44%, and 63% lower muscle insulin sensitivity (IS), metabolic flexibility (P < 0.01) and AT IS (P < 0.05), 73% and 31% higher HCL (P < 0.05), and DSAT-thickness (P < 0.001), but similar hepatic IS compared with CON. Mitochondrial efficiency was ~22% lower in SSAT and DSAT of T2D patients (P < 0.001) and ~8% lower in SSAT vs DSAT (P < 0.05). In both fat depots, mitochondrial coupling correlated positively with muscle IS and metabolic flexibility (r ≥ 0.40; P < 0.05), proton leak correlated positively (r ≥ 0.51; P < 0.01) and oxidative capacity negatively (r ≤ -0.47; P < 0.05) with fasting free fatty acids (FFA). Metabolic flexibility correlated positively with SAT-oxidative capacity (r ≥ 0.48; P < 0.05) and negatively with DSAT-thickness (r = -0.48; P < 0.05). DSAT-thickness correlated negatively with mitochondrial coupling in both depots (r ≤ -0.50; P < 0.01) and muscle IS (r = -0.59; P < 0.01), positively with FFA during clamp (r = 0.63; P < 0.001) and HCL (r = 0.49; P < 0.01). CONCLUSIONS Impaired mitochondrial function, insulin resistance, and DSAT expansion are AT abnormalities in recent-onset T2D that might promote whole-body insulin resistance and increased substrate flux to the liver.
Collapse
Affiliation(s)
- Kálmán Bódis
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Tomas Jelenik
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Jesper Lundbom
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Daniel F Markgraf
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Alexander Strom
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Oana-Patricia Zaharia
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Yanislava Karusheva
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Volker Burkart
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Karsten Müssig
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Yuliya Kupriyanova
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Meriem Ouni
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | | | - Jong-Hee Hwang
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Dan Ziegler
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Annette Schürmann
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - Michael Roden
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Julia Szendroedi
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Correspondence: Dr. Julia Szendroedi, PhD, Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany, c/o Auf’m Hennekamp 65, 40225 Düsseldorf, Germany. E-mail:
| | | |
Collapse
|
20
|
|
21
|
Li Y, Ma Q, Li P, Wang J, Wang M, Fan Y, Wang T, Wang C, Wang T, Zhao B. Proteomics reveals different pathological processes of adipose tissue, liver, and skeletal muscle under insulin resistance. J Cell Physiol 2020; 235:6441-6461. [PMID: 32115712 DOI: 10.1002/jcp.29658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 02/12/2020] [Indexed: 12/17/2022]
Abstract
Type 2 diabetes mellitus is the most common type of diabetes, and insulin resistance (IR) is its core pathological mechanism. Proteomics is an ingenious and promising Omics technology that can comprehensively describe the global protein expression profiling of body or specific tissue, and is widely applied to the study of molecular mechanisms of diseases. In this paper, we focused on insulin target organs: adipose tissue, liver, and skeletal muscle, and analyzed the different pathological processes of IR in these three tissues based on proteomics research. By literature studies, we proposed that the main pathological processes of IR among target organs were diverse, which showed unique characteristics and focuses. We further summarized the differential proteins in target organs which were verified to be related to IR, and discussed the proteins that may play key roles in the emphasized pathological processes, aiming at discovering potentially specific differential proteins of IR, and providing new ideas for pathological mechanism research of IR.
Collapse
Affiliation(s)
- Yaqi Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Quantao Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Pengfei Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jingkang Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Min Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanyuan Fan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Tieshan Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chunguo Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ting Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Baosheng Zhao
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
22
|
Takematsu E, Spencer A, Auster J, Chen PC, Graham A, Martin P, Baker AB. Genome wide analysis of gene expression changes in skin from patients with type 2 diabetes. PLoS One 2020; 15:e0225267. [PMID: 32084158 PMCID: PMC7034863 DOI: 10.1371/journal.pone.0225267] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/31/2019] [Indexed: 12/15/2022] Open
Abstract
Non-healing chronic ulcers are a serious complication of diabetes and are a major healthcare problem. While a host of treatments have been explored to heal or prevent these ulcers from forming, these treatments have not been found to be consistently effective in clinical trials. An understanding of the changes in gene expression in the skin of diabetic patients may provide insight into the processes and mechanisms that precede the formation of non-healing ulcers. In this study, we investigated genome wide changes in gene expression in skin between patients with type 2 diabetes and non-diabetic patients using next generation sequencing. We compared the gene expression in skin samples taken from 27 patients (13 with type 2 diabetes and 14 non-diabetic). This information may be useful in identifying the causal factors and potential therapeutic targets for the prevention and treatment of diabetic related diseases.
Collapse
Affiliation(s)
- Eri Takematsu
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - Adrianne Spencer
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - Jeff Auster
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - Po-Chih Chen
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
| | - Annette Graham
- Department of Biological and Biomedical Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Scotland, United Kingdom
| | - Patricia Martin
- Department of Biological and Biomedical Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Scotland, United Kingdom
| | - Aaron B. Baker
- University of Texas at Austin, Department of Biomedical Engineering, Austin, TX
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX
- The Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX
- Institute for Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, TX
- * E-mail:
| |
Collapse
|
23
|
Heinonen S, Jokinen R, Rissanen A, Pietiläinen KH. White adipose tissue mitochondrial metabolism in health and in obesity. Obes Rev 2020; 21:e12958. [PMID: 31777187 DOI: 10.1111/obr.12958] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 08/27/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022]
Abstract
White adipose tissue is one of the largest organs of the body. It plays a key role in whole-body energy status and metabolism; it not only stores excess energy but also secretes various hormones and metabolites to regulate body energy balance. Healthy adipose tissue capable of expanding is needed for metabolic well-being and to prevent accumulation of triglycerides to other organs. Mitochondria govern several important functions in the adipose tissue. We review the derangements of mitochondrial function in white adipose tissue in the obese state. Downregulation of mitochondrial function or biogenesis in the white adipose tissue is a central driver for obesity-associated metabolic diseases. Mitochondrial functions compromised in obesity include oxidative functions and renewal and enlargement of the adipose tissue through recruitment and differentiation of adipocyte progenitor cells. These changes adversely affect whole-body metabolic health. Dysfunction of the white adipose tissue mitochondria in obesity has long-term consequences for the metabolism of adipose tissue and the whole body. Understanding the pathways behind mitochondrial dysfunction may help reveal targets for pharmacological or nutritional interventions that enhance mitochondrial biogenesis or function in adipose tissue.
Collapse
Affiliation(s)
- Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Riikka Jokinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Aila Rissanen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Psychiatry, Helsinki University Hospital, Helsinki, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
24
|
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: 1.6] [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
|
25
|
Trojnar M, Patro-Małysza J, Kimber-Trojnar Ż, Leszczyńska-Gorzelak B, Mosiewicz J. Associations between Fatty Acid-Binding Protein 4⁻A Proinflammatory Adipokine and Insulin Resistance, Gestational and Type 2 Diabetes Mellitus. Cells 2019; 8:cells8030227. [PMID: 30857223 PMCID: PMC6468522 DOI: 10.3390/cells8030227] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 03/03/2019] [Accepted: 03/03/2019] [Indexed: 12/12/2022] Open
Abstract
There is ample scientific evidence to suggest a link between the fatty acid-binding protein 4 (FABP4) and insulin resistance, gestational (GDM), and type 2 (T2DM) diabetes mellitus. This novel proinflammatory adipokine is engaged in the regulation of lipid metabolism at the cellular level. The molecule takes part in lipid oxidation, the regulation of transcription as well as the synthesis of membranes. An involvement of FABP4 in the pathogenesis of obesity and insulin resistance seems to be mediated via FABP4-dependent peroxisome proliferator-activated receptor γ (PPARγ) inhibition. A considerable number of studies have shown that plasma concentrations of FABP4 is increased in obesity and T2DM, and that circulating FABP4 levels are correlated with certain clinical parameters, such as body mass index, insulin resistance, and dyslipidemia. Since plasma-circulating FABP4 has the potential to modulate the function of several types of cells, it appears to be of extreme interest to try to develop potential therapeutic strategies targeting the pathogenesis of metabolic diseases in this respect. In this manuscript, representing a detailed review of the literature on FABP4 and the abovementioned metabolic disorders, various mechanisms of the interaction of FABP4 with insulin signaling pathways are thoroughly discussed. Clinical aspects of insulin resistance in diabetic patients, including women diagnosed with GDM, are analyzed as well.
Collapse
Affiliation(s)
- Marcin Trojnar
- Chair and Department of Internal Medicine, Medical University of Lublin, 20-081 Lublin, Poland.
| | - Jolanta Patro-Małysza
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland.
| | - Żaneta Kimber-Trojnar
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland.
| | | | - Jerzy Mosiewicz
- Chair and Department of Internal Medicine, Medical University of Lublin, 20-081 Lublin, Poland.
| |
Collapse
|
26
|
Kras KA, Langlais PR, Hoffman N, Roust LR, Benjamin TR, De Filippis EA, Dinu V, Katsanos CS. Obesity modifies the stoichiometry of mitochondrial proteins in a way that is distinct to the subcellular localization of the mitochondria in skeletal muscle. Metabolism 2018; 89:18-26. [PMID: 30253140 PMCID: PMC6221946 DOI: 10.1016/j.metabol.2018.09.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/01/2018] [Accepted: 09/19/2018] [Indexed: 01/31/2023]
Abstract
BACKGROUND Skeletal muscle mitochondrial content and function appear to be altered in obesity. Mitochondria in muscle are found in well-defined regions within cells, and they are arranged in a way that form distinct subpopulations of subsarcolemmal (SS) and intermyofibrillar (IMF) mitochondria. We sought to investigate differences in the proteomes of SS and IMF mitochondria between lean subjects and subjects with obesity. METHODS We performed comparative proteomic analyses on SS and IMF mitochondria isolated from muscle samples obtained from lean subjects and subjects with obesity. Mitochondria were isolated using differential centrifugation, and proteins were subjected to label-free quantitative tandem mass spectrometry analyses. Collected data were evaluated for abundance of mitochondrial proteins using spectral counting. The Reactome pathway database was used to determine metabolic pathways that are altered in obesity. RESULTS Among proteins, 73 and 41 proteins showed different (mostly lower) expression in subjects with obesity in the SS and IMF mitochondria, respectively (false discovery rate-adjusted P ≤ 0.05). We specifically found an increase in proteins forming the tricarboxylic acid cycle and electron transport chain (ETC) complex II, but a decrease in proteins forming protein complexes I and III of the ETC and adenosine triphosphate (ATP) synthase in subjects with obesity in the IMF, but not SS, mitochondria. Obesity was associated with differential effects on metabolic pathways linked to protein translation in the SS mitochondria and ATP formation in the IMF mitochondria. CONCLUSIONS Obesity alters the expression of mitochondrial proteins regulating key metabolic processes in skeletal muscle, and these effects are distinct to mitochondrial subpopulations located in different regions of the muscle fibers. TRIAL REGISTRATION ClinicalTrials.gov (NCT01824173).
Collapse
Affiliation(s)
- Katon A Kras
- Center for Metabolic and Vascular Biology, Arizona State University, Scottsdale, AZ 85259, United States of America
| | - Paul R Langlais
- College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, United States of America
| | - Nyssa Hoffman
- Center for Metabolic and Vascular Biology, Arizona State University, Scottsdale, AZ 85259, United States of America
| | - Lori R Roust
- College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, United States of America
| | - Tonya R Benjamin
- College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, United States of America
| | - Elena A De Filippis
- College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, United States of America
| | - Valentin Dinu
- Department of Biomedical Informatics, Arizona State University, Scottsdale, AZ 85259, United States of America
| | - Christos S Katsanos
- Center for Metabolic and Vascular Biology, Arizona State University, Scottsdale, AZ 85259, United States of America; College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ 85259, United States of America.
| |
Collapse
|
27
|
Bódis K, Roden M. Energy metabolism of white adipose tissue and insulin resistance in humans. Eur J Clin Invest 2018; 48:e13017. [PMID: 30107041 DOI: 10.1111/eci.13017] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 07/22/2018] [Accepted: 08/12/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Insulin resistance not only occurs in obesity, but also in lipodystrophy. Although adipose tissue mass affects metabolic fluxes and participates in interorgan crosstalk, the role of energy metabolism within white adipose tissue for insulin resistance is less clear. MATERIALS AND METHODS A Medline search identified in vivo studies in humans on energy and lipid metabolism in subcutaneous (SAT) and visceral adipose tissue (VAT). Studies in adipocyte cultures and transgenic animal models were included for the better understanding of the link between abnormal energy metabolism in adipose tissue and insulin resistance. RESULTS The current literature indicates that higher lipolysis and lower lipogenesis in VAT compared to SAT enhance portal delivery of lipid metabolites (glycerol and fatty acids) to the liver. Thus, the lower lipolysis and higher lipogenesis in SAT favour storage of excess lipids and allow for protection of insulin-sensitive tissues from lipotoxic effects. In insulin-resistant humans, enhanced lipolysis and impaired lipogenesis in adipose tissue lead to release of cytokines and lipid metabolites, ultimately promoting insulin resistance. Adipose tissue of insulin-resistant humans also displays lower expression of proteins involved in mitochondrial function. In turn, this leads to lower availability of mitochondria-derived energy sources for lipogenesis in adipose tissue. CONCLUSIONS Abnormal mitochondrial function in human white adipose tissue likely contributes to the secretion of lipid metabolites and lactate, which are linked to insulin resistance in peripheral tissues. However, the relevance of adipose tissue energy metabolism for the regulation of human insulin sensitivity remains to be further elucidated.
Collapse
Affiliation(s)
- Kálmán Bódis
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Michael Roden
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
28
|
Masood A, Benabdelkamel H, Alfadda AA. Obesity Proteomics: An Update on the Strategies and Tools Employed in the Study of Human Obesity. High Throughput 2018; 7:ht7030027. [PMID: 30213114 PMCID: PMC6164994 DOI: 10.3390/ht7030027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/30/2018] [Accepted: 09/10/2018] [Indexed: 02/07/2023] Open
Abstract
Proteomics has become one of the most important disciplines for characterizing cellular protein composition, building functional linkages between protein molecules, and providing insight into the mechanisms of biological processes in a high-throughput manner. Mass spectrometry-based proteomic advances have made it possible to study human diseases, including obesity, through the identification and biochemical characterization of alterations in proteins that are associated with it and its comorbidities. A sizeable number of proteomic studies have used the combination of large-scale separation techniques, such as high-resolution two-dimensional gel electrophoresis or liquid chromatography in combination with mass spectrometry, for high-throughput protein identification. These studies have applied proteomics to comprehensive biochemical profiling and comparison studies while using different tissues and biological fluids from patients to demonstrate the physiological or pathological adaptations within their proteomes. Further investigations into these proteome-wide alterations will enable us to not only understand the disease pathophysiology, but also to determine signature proteins that can serve as biomarkers for obesity and related diseases. This review examines the different proteomic techniques used to study human obesity and discusses its successful applications along with its technical limitations.
Collapse
Affiliation(s)
- Afshan Masood
- Obesity Research Center, College of Medicine, King Saud University, P.O. Box 2925 (98), Riyadh 11461, Saudi Arabia.
| | - Hicham Benabdelkamel
- Obesity Research Center, College of Medicine, King Saud University, P.O. Box 2925 (98), Riyadh 11461, Saudi Arabia.
| | - Assim A Alfadda
- Obesity Research Center, College of Medicine, King Saud University, P.O. Box 2925 (98), Riyadh 11461, Saudi Arabia.
- Department of Medicine, College of Medicine, King Saud University, P.O. Box 2925 (38), Riyadh 11461, Saudi Arabia.
| |
Collapse
|
29
|
A Large Multiethnic Genome-Wide Association Study of Adult Body Mass Index Identifies Novel Loci. Genetics 2018; 210:499-515. [PMID: 30108127 DOI: 10.1534/genetics.118.301479] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/08/2018] [Indexed: 12/31/2022] Open
Abstract
Body mass index (BMI), a proxy measure for obesity, is determined by both environmental (including ethnicity, age, and sex) and genetic factors, with > 400 BMI-associated loci identified to date. However, the impact, interplay, and underlying biological mechanisms among BMI, environment, genetics, and ancestry are not completely understood. To further examine these relationships, we utilized 427,509 calendar year-averaged BMI measurements from 100,418 adults from the single large multiethnic Genetic Epidemiology Research on Adult Health and Aging (GERA) cohort. We observed substantial independent ancestry and nationality differences, including ancestry principal component interactions and nonlinear effects. To increase the list of BMI-associated variants before assessing other differences, we conducted a genome-wide association study (GWAS) in GERA, with replication in the Genetic Investigation of Anthropomorphic Traits (GIANT) consortium combined with the UK Biobank (UKB), followed by GWAS in GERA combined with GIANT, with replication in the UKB. We discovered 30 novel independent BMI loci (P < 5.0 × 10-8) that replicated. We then assessed the proportion of BMI variance explained by sex in the UKB using previously identified loci compared to previously and newly identified loci and found slight increases: from 3.0 to 3.3% for males and from 2.7 to 3.0% for females. Further, the variance explained by previously and newly identified variants decreased with increasing age in the GERA and UKB cohorts, echoed in the variance explained by the entire genome, which also showed gene-age interaction effects. Finally, we conducted a tissue expression QTL enrichment analysis, which revealed that GWAS BMI-associated variants were enriched in the cerebellum, consistent with prior work in humans and mice.
Collapse
|
30
|
Livadariu R, Timofte D, Trifan A, Danila R, Ionescu L, Sîngeap A, Ciobanu D. VITAMIN D DEFICIENCY, A NONINVASIVE MARKER OF STEATOHEPATITIS IN PATIENTS WITH OBESITY AND BIOPSY PROVEN NONALCOHOLIC FATTY LIVER DISEASE. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2018; 14:76-84. [PMID: 31149239 PMCID: PMC6516593 DOI: 10.4183/aeb.2018.76] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
CONTEXT Nonalcoholic fatty liver disease (NAFLD) includes simple steatosis, steatohepatitis (NASH) which can evolve with progressive fibrosis, cirrhosis and hepatocellular carcinoma. As liver biopsy cannot be used as a screening method, noninvasive markers are needed. OBJECTIVE The aim of this study was to test if there is a significant association between vitamin D deficit and the severity of NAFLD. DESIGN The patients were divided into two groups (vitamin D insufficiency/deficiency) and statistical analyses were performed on the correlation of clinical and biochemical characteristics with histopathological hepatic changes. SUBJECTS AND METHODS We prospectively studied 64 obese patients referred for bariatric surgery between 2014 and 2016 to our Surgical Unit. Anthropometric, clinical measurements, general and specific biological balance were noted. NAFLD diagnosis and activity score (NAS) were evaluated on liver biopsies. RESULTS Increased serum fibrinogen was correlated with NASH (p=0.005) and higher NAS grade. T2DM was positively correlated with liver fibrosis (p=0.002). 84.37% of the patients had vitamin D deficit and 15.62% were vitamin D insufficient. Lobular inflammation correlated with vitamin D deficit (p=0.040). Fibrosis (p=0.050) and steatohepatitis (p=0.032) were independent predictors of low vitamin D concentration. CONCLUSIONS Vitamin D status in conjunction with other parameters - such as T2DM - or serum biomarkers - namely fibrinogen level and PCR level - may point out the aggressive forms of NAFLD and the need for liver biopsy for appropriate management.
Collapse
Affiliation(s)
- R. Livadariu
- “Gr. T. Popa” University, Dept. of Surgery, Iasi, Romania
- “Gr. T. Popa” University, 3 Surgical Clinic, Iasi, Romania
| | - D. Timofte
- “Gr. T. Popa” University, Dept. of Surgery, Iasi, Romania
- “Gr. T. Popa” University, 3 Surgical Clinic, Iasi, Romania
| | - A. Trifan
- “Gr. T. Popa” University, Dept. of Gastroenterology, Iasi, Romania
- “Gr. T. Popa” University, Institute of Gastroenterology and Hepatology, Iasi, Romania
| | - R. Danila
- “Gr. T. Popa” University, Dept. of Surgery, Iasi, Romania
- “Gr. T. Popa” University, 3 Surgical Clinic, Iasi, Romania
| | - L. Ionescu
- “Gr. T. Popa” University, Dept. of Surgery, Iasi, Romania
- “Gr. T. Popa” University, 3 Surgical Clinic, Iasi, Romania
| | - A.M. Sîngeap
- “Gr. T. Popa” University, Dept. of Gastroenterology, Iasi, Romania
- “Gr. T. Popa” University, Institute of Gastroenterology and Hepatology, Iasi, Romania
| | - D. Ciobanu
- “Gr. T. Popa” University, Dept. of Pathology, “St. Spiridon” Emergency Hospital, Iasi, Romania
- “Gr. T. Popa” University, Dept. of Pathology, Iasi, Romania
| |
Collapse
|
31
|
Xie X, Sinha S, Yi Z, Langlais PR, Madan M, Bowen BP, Willis W, Meyer C. Role of adipocyte mitochondria in inflammation, lipemia and insulin sensitivity in humans: effects of pioglitazone treatment. Int J Obes (Lond) 2017; 42:ijo2017192. [PMID: 29087390 PMCID: PMC6021211 DOI: 10.1038/ijo.2017.192] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 07/19/2017] [Accepted: 08/02/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND/OBJECTIVES To gain further insight into the role of adipocyte mitochondria in systemic lipid metabolism, inflammation and insulin sensitivity in humans and to provide a better understanding of the mechanisms of action of the peroxisome proliferator-activated receptor gamma agonist pioglitazone. SUBJECTS/METHODS Mitochondrial DNA (mtDNA) copy number, mitochondrial distribution, mitochondrial and overall cellular protein abundances as well as intrinsic mitochondrial function of subcutaneous adipocytes were assessed by real-time quantitative PCR, MitoTracker staining, global proteomics analyses and NADH cytochrome c reductase activity in insulin-sensitive, normal-glucose-tolerant (NGT) individuals and age, gender, adiposity-matched insulin-resistant individuals with abnormal glucose tolerant (AGT) before and after 3 months of pioglitazone treatment. RESULTS mtDNA copy number/adipocyte and mtDNA copy number/adipocyte volume were ~55% and ~4-fold lower in AGT than in NGT, respectively, and correlated positively with the M-value of euglycemic clamps and high-density lipoprotein, and negatively with fasting plasma triglyceride, tumor necrosis factor-α and interleukin-6 levels in the entire cohort. mtDNA copy number/adipocyte volume also correlated positively with plasma adiponectin. Pioglitazone, which improved insulin sensitivity, plasma lipids and inflammation, increased the mitochondrial copy number, and led to a redistribution of mitochondria from a punctate to a more reticular pattern as observed in NGT. This was accompanied by disproportionately increased abundances of mitochondrial proteins, including those involved in fat oxidation and triglyceride synthesis. Pioglitazone also increased the abundance of collagen VI and decreased the abundance of cytoskeletal proteins. NADH cytochrome c reductase activity of isolated adipocyte mitochondria was similar in AGT and NGT and unaltered by pioglitazone. CONCLUSIONS Adipocyte mitochondria are deficient in insulin-resistant individuals and correlate with systemic lipid metabolism, inflammation and insulin sensitivity. Pioglitazone induces mitochondrial biogenesis and reorganization as well as the synthesis of mitochondrial proteins including those critical for lipid metabolism. It also alters extracellular matrix and cytoskeletal proteins. The intrinsic function of adipocyte mitochondria appears unaffected in insulin resistance and by pioglitazone.International Journal of Obesity advance online publication, 31 October 2017; doi:10.1038/ijo.2017.192.
Collapse
Affiliation(s)
- X Xie
- Center for Metabolic Biology, Arizona State University, Tempe, AZ, USA
| | - S Sinha
- Center for Metabolic Biology, Arizona State University, Tempe, AZ, USA
| | - Z Yi
- Center for Metabolic Biology, Arizona State University, Tempe, AZ, USA
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, MI, USA
| | - PR Langlais
- Center for Metabolic Biology, Arizona State University, Tempe, AZ, USA
| | - M Madan
- Center for Metabolic Biology, Arizona State University, Tempe, AZ, USA
| | - BP Bowen
- Center for Metabolic Biology, Arizona State University, Tempe, AZ, USA
| | - W Willis
- Center for Metabolic Biology, Arizona State University, Tempe, AZ, USA
| | - C Meyer
- Center for Metabolic Biology, Arizona State University, Tempe, AZ, USA
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, FL, USA
| |
Collapse
|
32
|
Gileles-Hillel A, Almendros I, Khalyfa A, Nigdelioglu R, Qiao Z, Hamanaka RB, Mutlu GM, Akbarpour M, Gozal D. Prolonged Exposures to Intermittent Hypoxia Promote Visceral White Adipose Tissue Inflammation in a Murine Model of Severe Sleep Apnea: Effect of Normoxic Recovery. Sleep 2017; 40:2731734. [PMID: 28329220 DOI: 10.1093/sleep/zsw074] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Study Objective Increased visceral white adipose tissue (vWAT) mass results in infiltration of inflammatory macrophages that drive inflammation and insulin resistance. Patients with obstructive sleep apnea (OSA) suffer from increased prevalence of obesity, insulin resistance, and metabolic syndrome. Murine models of intermittent hypoxia (IH) mimicking moderate-severe OSA manifest insulin resistance following short-term IH. We examined in mice the effect of long-term IH on the inflammatory cellular changes within vWAT and the potential effect of normoxic recovery (IH-R). Methods Male C57BL/6J mice were subjected to IH for 20 weeks, and a subset was allowed to recover in room air (RA) for 6 or 12 weeks (IH-R). Stromal vascular fraction was isolated from epididymal vWAT and mesenteric vWAT depots, and single-cell suspensions were prepared for flow cytometry analyses, reactive oxygen species (ROS), and metabolic assays. Results IH reduced body weight and vWAT mass and IH-R resulted in catch-up weight and vWAT mass. IH-exposed vWAT exhibited increased macrophage counts (ATMs) that were only partially improved in IH-R. IH also caused a proinflammatory shift in ATMs (increased Ly6c(hi)(+) and CD36(+) ATMs). These changes were accompanied by increased vWAT insulin resistance with only partial improvements in IH-R. In addition, ATMs exhibited increased ROS production, altered metabolism, and changes in electron transport chain, which were only partially improved in IH-R. Conclusion Prolonged exposures to IH during the sleep period induce pronounced vWAT inflammation and insulin resistance despite concomitant vWAT mass reductions. These changes are only partially reversible after 3 months of normoxic recovery. Thus, long-lasting OSA may preclude complete reversibility of metabolic changes.
Collapse
Affiliation(s)
- Alex Gileles-Hillel
- Sections of Pediatric Sleep Medicine and Pulmonology, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Isaac Almendros
- Sections of Pediatric Sleep Medicine and Pulmonology, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Abdelnaby Khalyfa
- Sections of Pediatric Sleep Medicine and Pulmonology, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Recep Nigdelioglu
- Department of Medicine, Section of Pulmonary and Critical Care, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Zhuanhong Qiao
- Sections of Pediatric Sleep Medicine and Pulmonology, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Robert B Hamanaka
- Department of Medicine, Section of Pulmonary and Critical Care, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Gökhan M Mutlu
- Department of Medicine, Section of Pulmonary and Critical Care, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Mahzad Akbarpour
- Sections of Pediatric Sleep Medicine and Pulmonology, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - David Gozal
- Sections of Pediatric Sleep Medicine and Pulmonology, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| |
Collapse
|