1
|
Ruíz-Uribe M, Enríquez-Schmidt J, Monrroy-Uarac M, Mautner-Molina C, Kalazich-Rosales M, Muñoz M, Fuentes-Leal F, Cárcamo-Ibaceta C, Fazakerley DJ, Larance M, Ehrenfeld P, Martínez-Huenchullán S. Moderate-Intensity Constant and High-Intensity Interval Training Confer Differential Metabolic Benefits in Skeletal Muscle, White Adipose Tissue, and Liver of Candidates to Undergo Bariatric Surgery. J Clin Med 2024; 13:3273. [PMID: 38892984 PMCID: PMC11172953 DOI: 10.3390/jcm13113273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/24/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Background/Objectives: Bariatric surgery candidates require presurgical physical training, therefore, we compared the metabolic effects of a constant moderate-intensity training program (MICT) vs. a high-intensity interval training (HIIT) in this population. Methods: Seventeen participants performed MICT (n = 9, intensity of 50% of heart rate reserve (HRR) and/or 4-5/10 subjective sensation of effort (SSE)) or HIIT (n = 8, 6 cycles of 2.5 min at 80% of the HRR and/or 7-8/10 of SSE, interspersed by 6 cycles of active rest at 20% of the FCR) for 10 sessions for 4 weeks. After training, tissue samples (skeletal muscle, adipose tissue, and liver) were extracted, and protein levels of adiponectin, GLUT4, PGC1α, phospho-AMPK/AMPK, collagen 1 and TGFβ1 were measured. Results: Participants who performed MICT showed higher protein levels of PGC-1α in skeletal muscle samples (1.1 ± 0.27 vs. 0.7 ± 0.4-fold change, p < 0.05). In the liver samples of the people who performed HIIT, lower protein levels of phospho-AMPK/AMPK (1.0 ± 0.37 vs. 0.52 ± 0.22-fold change), PGC-1α (1.0 ± 0.18 vs. 0.69 ± 0.15-fold change), and collagen 1 (1.0 ± 0.26 vs. 0.59 ± 0.28-fold change) were observed (all p < 0.05). In subcutaneous adipose tissue, higher adiponectin levels were found only after HIIT training (1.1 ± 0.48 vs. 1.9 ± 0.69-fold change, p < 0.05). Conclusions: Our results show that both MICT and HIIT confer metabolic benefits in candidates undergoing bariatric surgery; however, most of these benefits have a program-specific fashion. Future studies should aim to elucidate the mechanisms behind these differences.
Collapse
Affiliation(s)
- Matías Ruíz-Uribe
- Cardiorespiratory and Metabolic Function Laboratory–Neyün, Valdivia 5090000, Chile;
| | - Javier Enríquez-Schmidt
- Exercise Physiology Laboratory, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile; (J.E.-S.); (M.M.-U.)
- Physical Therapy Unit, Locomotor Apparatus and Rehabilitation Institute, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Manuel Monrroy-Uarac
- Exercise Physiology Laboratory, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile; (J.E.-S.); (M.M.-U.)
- Physical Therapy Unit, Locomotor Apparatus and Rehabilitation Institute, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Camila Mautner-Molina
- Clínica Alemana de Valdivia, Valdivia 5090000, Chile; (C.M.-M.); (M.K.-R.); (M.M.); (F.F.-L.); (C.C.-I.)
| | - Mariana Kalazich-Rosales
- Clínica Alemana de Valdivia, Valdivia 5090000, Chile; (C.M.-M.); (M.K.-R.); (M.M.); (F.F.-L.); (C.C.-I.)
| | - Maximiliano Muñoz
- Clínica Alemana de Valdivia, Valdivia 5090000, Chile; (C.M.-M.); (M.K.-R.); (M.M.); (F.F.-L.); (C.C.-I.)
| | - Francisca Fuentes-Leal
- Clínica Alemana de Valdivia, Valdivia 5090000, Chile; (C.M.-M.); (M.K.-R.); (M.M.); (F.F.-L.); (C.C.-I.)
| | - Carlos Cárcamo-Ibaceta
- Clínica Alemana de Valdivia, Valdivia 5090000, Chile; (C.M.-M.); (M.K.-R.); (M.M.); (F.F.-L.); (C.C.-I.)
- Surgery Institute, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Daniel J. Fazakerley
- Metabolic Research Laboratory, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 1TN, UK;
| | - Mark Larance
- Charles Perkins Centre and School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Pamela Ehrenfeld
- Cellular Pathology Laboratory, Anatomy, Histology, and Pathology Institute, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile;
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Sergio Martínez-Huenchullán
- Cardiorespiratory and Metabolic Function Laboratory–Neyün, Valdivia 5090000, Chile;
- Nephrology Division, School of Medicine, Universidad Austral de Chile, Valdivia 5090000, Chile
- School of Physical Therapy, Universidad San Sebastián, Valdivia 5090000, Chile
| |
Collapse
|
2
|
González-Domínguez Á, Belmonte T, González-Domínguez R. Childhood obesity, metabolic syndrome, and oxidative stress: microRNAs go on stage. Rev Endocr Metab Disord 2023; 24:1147-1164. [PMID: 37672200 PMCID: PMC10698091 DOI: 10.1007/s11154-023-09834-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/25/2023] [Indexed: 09/07/2023]
Abstract
The incidence of childhood obesity and metabolic syndrome has grown notably in the last years, becoming major public health burdens in developed countries. Nowadays, oxidative stress is well-recognized to be closely associated with the onset and progression of several obesity-related complications within the framework of a complex crosstalk involving other intertwined pathogenic events, such as inflammation, insulin disturbances, and dyslipidemia. Thus, understanding the molecular basis behind these oxidative dysregulations could provide new approaches for the diagnosis, prevention, and treatment of childhood obesity and associated disorders. In this respect, the transcriptomic characterization of miRNAs bares great potential because of their involvement in post-transcriptional modulation of genetic expression. Herein, we provide a comprehensive literature revision gathering state-of-the-art research into the association between childhood obesity, metabolic syndrome, and miRNAs. We put special emphasis on the potential role of miRNAs in modulating obesity-related pathogenic events, with particular focus on oxidative stress.
Collapse
Affiliation(s)
- Álvaro González-Domínguez
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Universidad de Cádiz, Cádiz, 11009, Spain.
| | - Thalía Belmonte
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
| | - Raúl González-Domínguez
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Universidad de Cádiz, Cádiz, 11009, Spain
| |
Collapse
|
3
|
Orioli L, Canouil M, Sawadogo K, Ning L, Deldicque L, Lause P, de Barsy M, Froguel P, Loumaye A, Deswysen Y, Navez B, Bonnefond A, Thissen JP. Identification of myokines susceptible to improve glucose homeostasis after bariatric surgery. Eur J Endocrinol 2023; 189:409-421. [PMID: 37638789 DOI: 10.1093/ejendo/lvad122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/20/2023] [Accepted: 08/03/2023] [Indexed: 08/29/2023]
Abstract
IMPORTANCE AND OBJECTIVE The identification of myokines susceptible to improve glucose homeostasis following bariatric surgery could lead to new therapeutic approaches for type 2 diabetes. METHODS Changes in the homeostasis model assessment (HOMA) test were assessed in patients before and 3 months after bariatric surgery. Changes in myokines expression and circulating levels were assessed using real-time quantitative polymerase chain reaction (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA). Myokines known to regulate glucose homeostasis were identified using literature (targeted study) and putative myokines using RNA-sequencing (untargeted study). A linear regression analysis adjusted for age and sex was used to search for associations between changes in the HOMA test and changes in myokines. RESULTS In the targeted study, brain-derived neurotrophic factor (BDNF) expression was upregulated (+30%, P = .006) while BDNF circulating levels were decreased (-12%, P = .001). Upregulated BDNF expression was associated with decreased HOMA of insulin resistance (HOMA-IR) (adjusted estimate [95% confidence interval {CI}]: -0.51 [-0.88 to -0.13], P = .010). Decreased BDNF serum levels were associated with decreased HOMA of beta-cell function (HOMA-B) (adjusted estimate [95% CI] = 0.002 [0.00002-0.0031], P = .046). In the untargeted study, upregulated putative myokines included XYLT1 (+64%, P < .001), LGR5 (+57, P< .001), and SPINK5 (+46%, P < .001). Upregulated LGR5 was associated with decreased HOMA-IR (adjusted estimate [95% CI] = -0.50 [-0.86 to -0.13], P = .009). Upregulated XYLT1 and SPINK5 were associated with increased HOMA of insulin sensitivity (HOMA-S) (respectively, adjusted estimate [95% CI] = 109.1 [28.5-189.8], P = .009 and 16.5 [0.87-32.19], P = .039). CONCLUSIONS Improved glucose homeostasis following bariatric surgery is associated with changes in myokines expression and circulating levels. In particular, upregulation of BDNF, XYLT1, SPINK5, and LGR5 is associated with improved insulin sensitivity. These results suggest that these myokines could contribute to improved glucose homeostasis following bariatric surgery. STUDY REGISTRATION NCT03341793 on ClinicalTrials.gov (https://clinicaltrials.gov/).
Collapse
Affiliation(s)
- Laura Orioli
- Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Mickaël Canouil
- Inserm U1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, 59000 Lille, France
- University of Lille, Lille University Hospital, 59000 Lille, France
| | - Kiswendsida Sawadogo
- Statistical Support Unit, King Albert II Cancer and Hematology Institute, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Lijiao Ning
- Inserm U1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, 59000 Lille, France
- University of Lille, Lille University Hospital, 59000 Lille, France
| | - Louise Deldicque
- Institute of NeuroScience, Université Catholique de Louvain, 1348 Louvain-La-Neuve, Belgium
| | - Pascale Lause
- Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Marie de Barsy
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Philippe Froguel
- Inserm U1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, 59000 Lille, France
- University of Lille, Lille University Hospital, 59000 Lille, France
- Department of Metabolism, Digestion, and Reproduction, Imperial College London, London SW7 2BX, United Kingdom
| | - Audrey Loumaye
- Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Yannick Deswysen
- Department of Oeso-gastro-duodenal and Bariatric Surgery, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Benoit Navez
- Department of Oeso-gastro-duodenal and Bariatric Surgery, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Amélie Bonnefond
- Inserm U1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, 59000 Lille, France
- University of Lille, Lille University Hospital, 59000 Lille, France
| | - Jean-Paul Thissen
- Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
4
|
Zheng P, Ma W, Gu Y, Wu H, Bian Z, Liu N, Yang D, Chen X. High-fat diet causes mitochondrial damage and downregulation of mitofusin-2 and optic atrophy-1 in multiple organs. J Clin Biochem Nutr 2023; 73:61-76. [PMID: 37534099 PMCID: PMC10390808 DOI: 10.3164/jcbn.22-73] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 02/19/2023] [Indexed: 08/04/2023] Open
Abstract
High-fat consumption promotes the development of obesity, which is associated with various chronic illnesses. Mitochondria are the energy factories of eukaryotic cells, maintaining self-stability through a fine-tuned quality-control network. In the present study, we evaluated high-fat diet (HFD)-induced changes in mitochondrial ultrastructure and dynamics protein expression in multiple organs. C57BL/6J male mice were fed HFD or normal diet (ND) for 24 weeks. Compared with ND-fed mice, HFD-fed mice exhibited increased body weight, cardiomyocyte enlargement, pulmonary fibrosis, hepatic steatosis, renal and splenic structural abnormalities. The cellular apoptosis of the heart, liver, and kidney increased. Cellular lipid droplet deposition and mitochondrial deformations were observed. The proteins related to mitochondrial biogenesis (TFAM), fission (DRP1), autophagy (LC3 and LC3-II: LC3-I ratio), and mitophagy (PINK1) presented different changes in different organs. The mitochondrial fusion regulators mitofusin-2 (MFN2) and optic atrophy-1 (OPA1) were consistently downregulated in multiple organs, even the spleen. TOMM20 and ATP5A protein were enhanced in the heart, skeletal muscle, and spleen, and attenuated in the kidney. These results indicated that high-fat feeding caused pathological changes in multiple organs, accompanied by mitochondrial ultrastructural damage, and MFN2 and OPA1 downregulation. The mitochondrial fusion proteins may become promising targets and/or markers for treating metabolic disease.
Collapse
Affiliation(s)
- Peng Zheng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Wenjing Ma
- Core Facility, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Yilu Gu
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Hengfang Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Zhiping Bian
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Nannan Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Di Yang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
- Core Facility, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Xiangjian Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| |
Collapse
|
5
|
Gu Y, Yu W, Qi M, Hu J, Jin Q, Wang X, Wang C, Chen Y, Yuan W. Identification and validation of hub genes and pathways associated with mitochondrial dysfunction in hypertrophy of ligamentum flavum. Front Genet 2023; 14:1117416. [PMID: 37234868 PMCID: PMC10206037 DOI: 10.3389/fgene.2023.1117416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/22/2023] [Indexed: 05/28/2023] Open
Abstract
Background: Lumbar spinal stenosis which can lead to irreversible neurologic damage and functional disability, is characterized by hypertrophy of ligamentum flavum (HLF). Recent studies have indicated that mitochondrial dysfunction may contribute to the development of HLF. However, the underlying mechanism is still unclear. Methods: The dataset GSE113212 was obtained from the Gene Expression Omnibus database, and the differentially expressed genes were identified. The intersection of DEGs and mitochondrial dysfunction-related genes were identified as mitochondrial dysfunction-related DEGs. Gene Ontology analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, and Gene Set Enrichment Analysis were performed. Protein-protein interaction network was constructed, and miRNAs and transcriptional factors of the hub genes were predicted via the miRNet database. Small molecule drugs targeted to these hub genes were predicted via PubChem. Immune infiltration analysis was performed to evaluate the infiltration level of immune cells and their correlation with the hub genes. In final, we measured the mitochondrial function and oxidative stress in vitro and verified the expression of hub genes by qPCR experiments. Results: In total, 43 genes were identified as MDRDEGs. These genes were mainly involved in cellular oxidation, catabolic processes, and the integrity of mitochondrial structure and function. The top hub genes were screened, including LONP1, TK2, SCO2, DBT, TFAM, MFN2. The most significant enriched pathways include cytokine-cytokine receptor interaction, focal adhesion, etc. Besides, SP1, PPARGC1A, YY1, MYC, PPARG, and STAT1 were predicted transcriptional factors of these hub genes. Additionally, increased immune infiltration was demonstrated in HLF, with a close correlation between hub genes and immune cells found. The mitochondrial dysfunction and the expression of hub genes were validated by evaluation of mitochondrial DNA, oxidative stress markers and quantitative real-time PCR. Conclusion: This study applied the integrative bioinformatics analysis and revealed the mitochondrial dysfunction-related key genes, regulatory pathways, TFs, miRNAs, and small molecules underlying the development of HLF, which improved the understanding of molecular mechanisms and the development of novel therapeutic targets for HLF.
Collapse
Affiliation(s)
- Yifei Gu
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Wenchao Yu
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Min Qi
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jinquan Hu
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Qianmei Jin
- Department of Rheumatology and Immunology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xinwei Wang
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chen Wang
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yu Chen
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Wen Yuan
- Department of Orthopaedics, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
6
|
Pérez-Rodríguez M, Huertas JR, Villalba JM, Casuso RA. Mitochondrial adaptations to calorie restriction and bariatric surgery in human skeletal muscle: a systematic review with meta-analysis. Metabolism 2023; 138:155336. [PMID: 36302454 DOI: 10.1016/j.metabol.2022.155336] [Citation(s) in RCA: 6] [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] [Received: 07/28/2022] [Revised: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 11/18/2022]
Abstract
OBJECTIVE We performed a meta-analysis to determine the changes induced by calorie restriction (CR) and bariatric surgery on human skeletal muscle mitochondria. METHODS A systematic search of Medline and Web of Science was conducted. Controlled trials exploring CR (≥14 days) and mitochondrial function and/or content assessment were included. Moreover, studies analyzing weight loss following gastric surgery were included for comparison purposes. Human muscle data from 28 studies assessing CR (520 muscle samples) and from 10 studies assessing bariatric surgery (155 muscle samples) were analyzed in a random effect meta-analysis with three a priori chosen covariates. MAIN RESULTS We report a decrease (p < 0.05) (mean (95 % CI)) in maximal mitochondrial state 3 respiration in response to CR (-0.44 (-0.85, -0.03)) but not in response to surgery (-0.33 (-1.18, 0.52)). No changes in mitochondrial content were reported after CR (-0.05 (-0.12, 0.13)) or in response to surgery (0.23 (-0.05, 0.52)). Moreover, data from CR subjects showed a reduction in complex IV (CIV) activity (-0.29 (-0.56, -0.03)) but not in CIV content (-0.21 (-0.63, 0.22)). Similar results were obtained when the length of the protocol, the initial body mass index, and the estimated energy deficit were included in the model as covariates. CONCLUSION The observation of reduced maximal mitochondrial state 3, uncoupled respiration, and CIV activity without altering mitochondrial content suggests that, in human skeletal muscle, CR mainly modulates intrinsic mitochondrial function.
Collapse
Affiliation(s)
- Miguel Pérez-Rodríguez
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, and ceiA3Campus of International Excellence in Agrifood, Spain
| | | | - José M Villalba
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, and ceiA3Campus of International Excellence in Agrifood, Spain
| | - Rafael A Casuso
- Department of Physiology, University of Granada, Spain; Department of Health Sciences, Universidad Loyola Andalucía, Spain.
| |
Collapse
|
7
|
Abad-Jiménez Z, Vezza T, López-Domènech S, Fernández-Reyes M, Canet F, Morillas C, Gómez-Abril SÁ, Bañuls C, Víctor VM, Rocha M. Impact of Roux-en-Y Gastric Bypass on Mitochondrial Biogenesis and Dynamics in Leukocytes of Obese Women. Antioxidants (Basel) 2022; 11:antiox11071302. [PMID: 35883794 PMCID: PMC9312345 DOI: 10.3390/antiox11071302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 02/05/2023] Open
Abstract
The chronic low-grade inflammation widely associated with obesity can lead to a prooxidant status that triggers mitochondrial dysfunction. To date, Roux-en-Y gastric bypass (RYGB) is considered the most effective strategy for obese patients. However, little is known about its molecular mechanisms. This interventional study aimed to investigate whether RYGB modulates oxidative stress, inflammation and mitochondrial dynamics in the leukocytes of 47 obese women at one year follow-up. We evaluated biochemical parameters and serum inflammatory cytokines -TNFα, IL6 and IL1β- to assess systemic status. Total superoxide production -dHe-, mitochondrial membrane potential -TMRM-, leucocyte protein expression of inflammation mediators -MCP1 and NF-kB-, antioxidant defence -GPX1-, mitochondrial regulation—PGC1α, TFAM, OXPHOS and MIEAP- and dynamics -MFN2, MNF1, OPA1, FIS1 and p-DRP1- were also determined. After RYGB, a significant reduction in superoxide and mitochondrial membrane potential was evident, while GPX1 content was significantly increased. Likewise, a marked upregulation of the transcription factors PGC1α and TFAM, complexes of the oxidative phosphorylation chain (I–V) and MIEAP and MFN1 was observed. We conclude that women undergoing RYGB benefit from an amelioration of their prooxidant and inflammatory status and an improvement in mitochondrial dynamics of their leukocytes, which is likely to have a positive effect on clinical outcome.
Collapse
Affiliation(s)
- Zaida Abad-Jiménez
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (T.V.); (S.L.-D.); (M.F.-R.); (F.C.); (C.M.); (C.B.)
| | - Teresa Vezza
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (T.V.); (S.L.-D.); (M.F.-R.); (F.C.); (C.M.); (C.B.)
| | - Sandra López-Domènech
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (T.V.); (S.L.-D.); (M.F.-R.); (F.C.); (C.M.); (C.B.)
| | - Meylin Fernández-Reyes
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (T.V.); (S.L.-D.); (M.F.-R.); (F.C.); (C.M.); (C.B.)
| | - Francisco Canet
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (T.V.); (S.L.-D.); (M.F.-R.); (F.C.); (C.M.); (C.B.)
| | - Carlos Morillas
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (T.V.); (S.L.-D.); (M.F.-R.); (F.C.); (C.M.); (C.B.)
| | - Segundo Ángel Gómez-Abril
- Department of General and Digestive System Surgery, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain;
- Department of Surgery, Faculty of Medicine and Dentistry, University of Valencia, Av. Blasco Ibáñez 13, 46010 Valencia, Spain
| | - Celia Bañuls
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (T.V.); (S.L.-D.); (M.F.-R.); (F.C.); (C.M.); (C.B.)
| | - Víctor M. Víctor
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (T.V.); (S.L.-D.); (M.F.-R.); (F.C.); (C.M.); (C.B.)
- CIBERehd-Department of Pharmacology, University of Valencia, Av. Blasco Ibáñez 13, 46010 Valencia, Spain
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
- Correspondence: (V.M.V.); (M.R.); Tel.: +34-96-318-91-32 (M.R.)
| | - Milagros Rocha
- Department of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (Z.A.-J.); (T.V.); (S.L.-D.); (M.F.-R.); (F.C.); (C.M.); (C.B.)
- CIBERehd-Department of Pharmacology, University of Valencia, Av. Blasco Ibáñez 13, 46010 Valencia, Spain
- Correspondence: (V.M.V.); (M.R.); Tel.: +34-96-318-91-32 (M.R.)
| |
Collapse
|
8
|
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
|
9
|
Rigalli JP, Theile D, Nilles J, Weiss J. Regulation of PXR Function by Coactivator and Corepressor Proteins: Ligand Binding Is Just the Beginning. Cells 2021; 10:cells10113137. [PMID: 34831358 PMCID: PMC8625645 DOI: 10.3390/cells10113137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 12/13/2022] Open
Abstract
The pregnane X receptor (PXR, NR1I2) is a nuclear receptor which exerts its regulatory function by heterodimerization with the retinoid-X-receptor α (RXRα, NR2B1) and binding to the promoter and enhancer regions of diverse target genes. PXR is involved in the regulation of drug metabolism and excretion, metabolic and immunological functions and cancer pathogenesis. PXR activity is strongly regulated by the association with coactivator and corepressor proteins. Coactivator proteins exhibit histone acetyltransferase or histone methyltransferase activity or associate with proteins having one of these activities, thus promoting chromatin decondensation and activation of the gene expression. On the contrary, corepressor proteins promote histone deacetylation and therefore favor chromatin condensation and repression of the gene expression. Several studies pointed to clear cell- and ligand-specific differences in the activation of PXR. In this article, we will review the critical role of coactivator and corepressor proteins as molecular determinants of the specificity of PXR-mediated effects. As already known for other nuclear receptors, understanding the complex mechanism of PXR activation in each cell type and under particular physiological and pathophysiological conditions may lead to the development of selective modulators with therapeutic potential.
Collapse
|
10
|
Fealy CE, Grevendonk L, Hoeks J, Hesselink MKC. Skeletal muscle mitochondrial network dynamics in metabolic disorders and aging. Trends Mol Med 2021; 27:1033-1044. [PMID: 34417125 DOI: 10.1016/j.molmed.2021.07.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/13/2021] [Accepted: 07/26/2021] [Indexed: 01/05/2023]
Abstract
With global demographics trending towards an aging population, the numbers of individuals with an age-associated loss of independence is increasing. A key contributing factor is loss of skeletal muscle mitochondrial, metabolic, and contractile function. Recent advances in imaging technologies have demonstrated the importance of mitochondrial morphology and dynamics in the pathogenesis of disease. In this review, we examine the evidence for altered mitochondrial dynamics as a mechanism in age and obesity-associated loss of skeletal muscle function, with a particular focus on the available human data. We highlight some of the areas where more data are needed to identify the specific mechanisms connecting mitochondrial morphology and skeletal muscle dysfunction.
Collapse
Affiliation(s)
- Ciarán E Fealy
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands; Department of Physical Education and Sport Sciences, University of Limerick, Castletroy, Limerick, Ireland
| | - Lotte Grevendonk
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Joris Hoeks
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Matthijs K C Hesselink
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
11
|
Axelrod CL, Fealy CE, Erickson ML, Davuluri G, Fujioka H, Dantas WS, Huang E, Pergola K, Mey JT, King WT, Mulya A, Hsia D, Burguera B, Tandler B, Hoppel CL, Kirwan JP. Lipids activate skeletal muscle mitochondrial fission and quality control networks to induce insulin resistance in humans. Metabolism 2021; 121:154803. [PMID: 34090870 PMCID: PMC8277749 DOI: 10.1016/j.metabol.2021.154803] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/10/2021] [Accepted: 05/31/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND AIMS A diminution in skeletal muscle mitochondrial function due to ectopic lipid accumulation and excess nutrient intake is thought to contribute to insulin resistance and the development of type 2 diabetes. However, the functional integrity of mitochondria in insulin-resistant skeletal muscle remains highly controversial. METHODS 19 healthy adults (age:28.4 ± 1.7 years; BMI:22.7 ± 0.3 kg/m2) received an overnight intravenous infusion of lipid (20% Intralipid) or saline followed by a hyperinsulinemic-euglycemic clamp to assess insulin sensitivity using a randomized crossover design. Skeletal muscle biopsies were obtained after the overnight lipid infusion to evaluate activation of mitochondrial dynamics proteins, ex-vivo mitochondrial membrane potential, ex-vivo oxidative phosphorylation and electron transfer capacity, and mitochondrial ultrastructure. RESULTS Overnight lipid infusion increased dynamin related protein 1 (DRP1) phosphorylation at serine 616 and PTEN-induced kinase 1 (PINK1) expression (P = 0.003 and P = 0.008, respectively) in skeletal muscle while reducing mitochondrial membrane potential (P = 0.042). The lipid infusion also increased mitochondrial-associated lipid droplet formation (P = 0.011), the number of dilated cristae, and the presence of autophagic vesicles without altering mitochondrial number or respiratory capacity. Additionally, lipid infusion suppressed peripheral glucose disposal (P = 0.004) and hepatic insulin sensitivity (P = 0.014). CONCLUSIONS These findings indicate that activation of mitochondrial fission and quality control occur early in the onset of insulin resistance in human skeletal muscle. Targeting mitochondrial dynamics and quality control represents a promising new pharmacological approach for treating insulin resistance and type 2 diabetes. CLINICAL TRIAL REGISTRATION NCT02697201, ClinicalTrials.gov.
Collapse
Affiliation(s)
- Christopher L Axelrod
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; Department of Translational Services, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Ciaran E Fealy
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Melissa L Erickson
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Gangarao Davuluri
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; Sarcopenia and Malnutrition Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Hisashi Fujioka
- Cryo-Electron Microscopy Core, Case Western Reserve University, Cleveland, OH 44109, USA; Center for Mitochondrial Diseases, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Wagner S Dantas
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Emily Huang
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Kathryn Pergola
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; Department of Translational Services, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Jacob T Mey
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - William T King
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; Department of Translational Services, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Anny Mulya
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Daniel Hsia
- Clinical Trials Unit, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | - Bartolome Burguera
- Endocrinology and Metabolism Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Bernard Tandler
- Center for Mitochondrial Diseases, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, OH 44106, USA
| | - Charles L Hoppel
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; Center for Mitochondrial Diseases, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44109, USA
| | - John P Kirwan
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA; Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
12
|
Slivka D, Dumke C, Hailes W, Ruby B. Impact of Hypoxic Exercise Recovery on Skeletal Muscle Glycogen and Gene Expression. High Alt Med Biol 2021; 22:300-307. [PMID: 34142871 DOI: 10.1089/ham.2021.0028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Slivka, Dustin, Charles Dumke, Walter Hailes, and Brent Ruby. Impact of hypoxic exercise recovery on skeletal muscle glycogen and gene expression. High Alt Med Biol. 22:300-307, 2021. Background: The impact of altitude during recovery from exercise is largely unknown. The purpose of this study was to determine the acute gene response and muscle glycogen re-synthesis after exercise when exposed to simulated high altitude during recovery. Materials and Methods: Twelve male participants (age, 25 ± 2 years; height, 178 ± 7 cm; weight, 78.8 ± 7.8 kg; VO2peak, 4.25 ± 0.59 l/min; Wpeak 307 ± 44 W; and body fat, 13.1% ± 1.2%) completed two trials (random order), which consisted of cycling for 90 minutes in laboratory conditions and then recovering for 6 hours in laboratory conditions (975 m; normoxia) or at a high simulated altitude (5,000 m; hypoxia). Results: Skeletal muscle biopsies from the vastus lateralis were obtained before exercise, after exercise, and 6 hours after exercise for the measurement of metabolic gene expression and muscle glycogen. Muscle glycogen decreased with exercise (61% ± 13%, p < 0.05) and increased with recovery (78% ± 35%, p < 0.05) with no difference between trials (p > 0.05). Hypoxia-inducible factor (HIF)-1α, HIF-2α, optic atrophy gene 1 (OPA-1), mitofusin 2 (MFN-2), and peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α) gene expression were suppressed after altitude exposure (p < 0.05), while mitochondrial fission 1 protein (FIS-1), phosphofructokinase (PFK), Cytochrome c oxidase (COX), and hexokinase (HK) were unaffected by altitude exposure (p > 0.05). Conclusions: High-altitude exposure during recovery from exercise inhibits gene expression associated with mitochondrial development without affecting muscle glycogen re-synthesis.
Collapse
Affiliation(s)
- Dustin Slivka
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Charles Dumke
- School of Integrative Physiology and Athletic Training, University of Montana, Missoula, Montana, USA
| | - Walter Hailes
- School of Integrative Physiology and Athletic Training, University of Montana, Missoula, Montana, USA
| | - Brent Ruby
- School of Integrative Physiology and Athletic Training, University of Montana, Missoula, Montana, USA
| |
Collapse
|
13
|
Garcia LA, Day SE, Coletta RL, Campos B, Benjamin TR, De Filippis E, Madura JA, Mandarino LJ, Roust LR, Coletta DK. Weight loss after Roux-En-Y gastric bypass surgery reveals skeletal muscle DNA methylation changes. Clin Epigenetics 2021; 13:100. [PMID: 33933146 PMCID: PMC8088644 DOI: 10.1186/s13148-021-01086-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/21/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The mechanisms of weight loss and metabolic improvements following bariatric surgery in skeletal muscle are not well known; however, epigenetic modifications are likely to contribute. The aim of our study was to investigate skeletal muscle DNA methylation after weight loss induced by Roux-en-Y gastric bypass (RYGB) surgery. Muscle biopsies were obtained basally from seven insulin-resistant obese (BMI > 40 kg/m2) female subjects (45.1 ± 3.6 years) pre- and 3-month post-surgery with euglycemic hyperinsulinemic clamps to assess insulin sensitivity. Four lean (BMI < 25 kg/m2) females (38.5 ± 5.8 years) served as controls. We performed reduced representation bisulfite sequencing next generation methylation on DNA isolated from the vastus lateralis muscle biopsies. RESULTS Global methylation was significantly higher in the pre- (32.97 ± 0.02%) and post-surgery (33.31 ± 0.02%) compared to the lean (30.46 ± 0.02%), P < 0.05. MethylSig analysis identified 117 differentially methylated cytosines (DMCs) that were significantly altered in the post- versus pre-surgery (Benjamini-Hochberg q < 0.05). In addition, 2978 DMCs were significantly altered in the pre-surgery obese versus the lean controls (Benjamini-Hochberg q < 0.05). For the post-surgery obese versus the lean controls, 2885 DMCs were altered (Benjamini-Hochberg q < 0.05). Seven post-surgery obese DMCs were normalized to levels similar to those observed in lean controls. Of these, 5 were within intergenic regions (chr11.68,968,018, chr16.73,100,688, chr5.174,115,531, chr5.1,831,958 and chr9.98,547,011) and the remaining two DMCs chr17.45,330,989 and chr14.105,353,824 were within in the integrin beta 3 (ITGB3) promoter and KIAA0284 exon, respectively. ITGB3 methylation was significantly decreased in the post-surgery (0.5 ± 0.5%) and lean controls (0 ± 0%) versus pre-surgery (13.6 ± 2.7%, P < 0.05). This decreased methylation post-surgery was associated with an increase in ITGB3 gene expression (fold change + 1.52, P = 0.0087). In addition, we showed that ITGB3 promoter methylation in vitro significantly suppressed transcriptional activity (P < 0.05). Transcription factor binding analysis for ITGB3 chr17.45,330,989 identified three putative transcription factor binding motifs; PAX-5, p53 and AP-2alphaA. CONCLUSIONS These results demonstrate that weight loss after RYGB alters the epigenome through DNA methylation. In particular, this study highlights ITGB3 as a novel gene that may contribute to the metabolic improvements observed post-surgery. Future additional studies are warranted to address the exact mechanism of ITGB3 in skeletal muscle.
Collapse
Affiliation(s)
- Luis A Garcia
- Department of Medicine, Division of Endocrinology, The University of Arizona College of Medicine, 1501 N. Campbell Ave, PO Box 245035, Tucson, AZ, 85724-5035, USA
| | - Samantha E Day
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - Richard L Coletta
- Department of Medicine, Division of Endocrinology, The University of Arizona College of Medicine, 1501 N. Campbell Ave, PO Box 245035, Tucson, AZ, 85724-5035, USA
| | - Baltazar Campos
- Department of Medicine, Division of Endocrinology, The University of Arizona College of Medicine, 1501 N. Campbell Ave, PO Box 245035, Tucson, AZ, 85724-5035, USA
| | - Tonya R Benjamin
- Department of Endocrinology, Metabolism and Diabetes, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Eleanna De Filippis
- Department of Endocrinology, Metabolism and Diabetes, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | | | - Lawrence J Mandarino
- Department of Medicine, Division of Endocrinology, The University of Arizona College of Medicine, 1501 N. Campbell Ave, PO Box 245035, Tucson, AZ, 85724-5035, USA
| | - Lori R Roust
- Department of Endocrinology, Metabolism and Diabetes, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Dawn K Coletta
- Department of Medicine, Division of Endocrinology, The University of Arizona College of Medicine, 1501 N. Campbell Ave, PO Box 245035, Tucson, AZ, 85724-5035, USA. .,Department of Physiology, The University of Arizona College of Medicine, Tucson, AZ, USA.
| |
Collapse
|
14
|
García-Roche M, Cañibe G, Casal A, Mattiauda DA, Ceriani M, Jasinsky A, Cassina A, Quijano C, Carriquiry M. Glucose and Fatty Acid Metabolism of Dairy Cows in a Total Mixed Ration or Pasture-Based System During Lactation. FRONTIERS IN ANIMAL SCIENCE 2021. [DOI: 10.3389/fanim.2021.622500] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
In this study, we explored mechanisms related to glucose and fatty acid metabolism in Holstein–Friesian multiparous dairy cows during lactation under two feeding strategies. From 0 to 180 days postpartum, cows were fed total mixed ration (TMR) ad libitum (non-grazing group, G0) or grazed Festuca arundinacea or Medicago sativa and were supplemented with 5.4 kg DM/d of an energy-protein concentrate (grazing group, G1). From 180 to 250 days postpartum, all cows grazed F. arundinacea and were supplemented with TMR. Plasma samples and liver biopsies were collected at −14, 35, 60, 110, 180, and 250 days in milk (DIM) for metabolite, hormone, gene expression, and western blot analysis. Our results showed increased levels of negative energy balance markers: plasma non-esterified fatty acids (NEFA), liver triglyceride and plasma β-hydroxybutyrate (BHB) (P < 0.01), triglyceride and β-hydroxybutyrate concentration were especially elevated for G1 cows. Also, hepatic mRNA expression of gluconeogenic enzymes was upregulated during early lactation (P < 0.05). In particular, methymalonyl-CoA mutase expression was increased for G0 cows (P < 0.05) while pyruvate carboxylase (PC) expression was increased for G1 cows (P < 0.05), suggesting differential gluconeogenic precursors for different feeding strategies. Phosphorylation of AMP-activated protein kinase was increased in early lactation vs. late lactation (P < 0.01) and negatively correlated with PC mRNA levels. The positive association of gluconeogenic genes with proliferator-activated receptor gamma coactivator 1-alpha (PPARGC1A) hepatic expression supported the importance of this transcription factor in glucose metabolism. The peroxisome proliferator-activated receptor alpha (PPARA) mRNA was increased during early lactation (P < 0.05), and was positively associated to PPARGC1A, carnitine palmitoyl-transferase 1, and hydroxymethylglutaryl-CoA synthase 2 (HMGCS2) mRNA expression. Alongside, hepatic mRNA expression of FABP was decreased for G1 vs. G0 cows (P < 0.05), possibly linked to impaired fatty acid transport and related to accumulation of liver triglycerides, evidencing G1 cows fail to adapt to the demands of early lactation. In sum, our results showed that metabolic adaptations related to early lactation negative energy balance can be affected by feeding strategy and might be regulated by the metabolic sensors AMPK, SIRT1, and coordinated by transcription factors PPARGC1A and PPARA.
Collapse
|
15
|
Emami MR, Jamshidi S, Zarezadeh M, Khorshidi M, Olang B, Sajadi Hezaveh Z, Sohouli M, Aryaeian N. Can vitamin E supplementation affect obesity indices? A systematic review and meta-analysis of twenty-four randomized controlled trials. Clin Nutr 2021; 40:3201-3209. [PMID: 33632535 DOI: 10.1016/j.clnu.2021.02.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Several mechanisms have been proposed for the effect of vitamin E on weight loss. Yet various interventional studies with wide ranges of doses and durations have reported contradictory results. METHODS Cochrane Library, PubMed, Scopus, and Embase databases were searched up to December 2020. Meta-analysis was performed using random-effect method. Effect size was presented as weighted mean difference (WMD) and 95% confidence interval (CI). Heterogeneity was evaluated using the I2 index. In order to identification of potential sources of heterogeneity, predefined subgroup and meta regression analyses was conducted. RESULTS A total of 24 studies with 33 data sets were included. There was no significant effect of vitamin E on weight (WMD: 0.15, 95% CI: -1.35 to 1.65, P = 0.847), body mass index (BMI) (WMD = 0.04, 95% CI: -0.29 to 0.37, P = 0.815), and waist circumference (WC) (WMD = -0.19 kg, 95% CI: -2.06 to 1.68, P = 0.842), respectively. However, subgroup analysis revealed that vitamin E supplementation in studies conducted on participants with normal BMI (18.5-24.9) had increasing impact on BMI (P = 0.047). CONCLUSION There was no significant effect of vitamin E supplementation on weight, BMI and WC. However, vitamin E supplementation might be associated with increasing BMI in people with normal BMI (18.5-24.9).
Collapse
Affiliation(s)
- Mohammad Reza Emami
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Sanaz Jamshidi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Meysam Zarezadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Masoud Khorshidi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran; Pediatric Gastroenterology, Hepatology and Nutrition Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Beheshteh Olang
- Pediatric Gastroenterology, Hepatology and Nutrition Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Community Medicine, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zohreh Sajadi Hezaveh
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Naheed Aryaeian
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Ferraz-Bannitz R, Welendorf CR, Coelho PO, Salgado W, Nonino CB, Beraldo RA, Foss-Freitas MC. Bariatric surgery can acutely modulate ER-stress and inflammation on subcutaneous adipose tissue in non-diabetic patients with obesity. Diabetol Metab Syndr 2021; 13:19. [PMID: 33593418 PMCID: PMC7887793 DOI: 10.1186/s13098-021-00623-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/02/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Bariatric surgery, especially Roux-en-Y gastric bypass (RYGB), is the most effective and durable treatment option for severe obesity. The mechanisms involving adipose tissue may be important to explain the effects of surgery. METHODS We aimed to identify the genetic signatures of adipose tissue in patients undergoing RYGB. We evaluated 13 obese, non-diabetic patients (mean age 37 years, 100% women, Body mass index (BMI) 42.2 kg/m2) one day before surgery, 3 and 6 months (M) after RYGB. RESULTS Analysis of gene expression in adipose tissue collected at surgery compared with samples collected at 3 M and 6 M Post-RYGB showed that interleukins [Interleukin 6, Tumor necrosis factor-α (TNF-α), and Monocyte chemoattractant protein-1(MCP1)] and endoplasmic reticulum stress (ERS) genes [Eukaryotic translation initiation factor 2 alpha kinase 3 (EIF2AK3) and Calreticulin (CALR)] decreased during the follow-up (P ≤ 0.01 for all). Otherwise, genes involved in energy homeostasis [Adiponectin and AMP-activated protein kinase (AMPK)], cellular response to oxidative stress [Sirtuin 1, Sirtuin 3, and Nuclear factor erythroid 2-related factor 2 (NRF2)], mitochondrial biogenesis [Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α)] and amino acids metabolism [General control nonderepressible 2 (GCN2)] increased from baseline to all other time points evaluated (P ≤ 0.01 for all). Also, expression of Peroxisome proliferator-activated receptor gamma (PPARϒ) (adipogenesis regulation) was significantly decreased after RYGB (P < 0.05). Additionally, we observed that PGC1α, SIRT1 and AMPK strongly correlated to BMI at 3 M (P ≤ 0.01 for all), as well as ADIPOQ and SIRT1 to BMI at 6 M (P ≤ 0.01 for all). CONCLUSIONS Our findings demonstrate that weight loss is associated with amelioration of inflammation and ERS and increased protection against oxidative stress in adipose tissue. These observations are strongly correlated with a decrease in BMI and essential genes that control cellular energy homeostasis, suggesting an adaptive process on a gene expression level during the caloric restriction and weight loss period after RYGB. Trial registration CAAE: 73,585,317.0.0000.5440.
Collapse
Affiliation(s)
- Rafael Ferraz-Bannitz
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo (USP), Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirao Preto, SP, 14049-900, Brazil.
| | - Caroline Rossi Welendorf
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo (USP), Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirao Preto, SP, 14049-900, Brazil
| | - Priscila Oliveira Coelho
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo (USP), Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirao Preto, SP, 14049-900, Brazil
| | - Wilson Salgado
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto, SP, Brazil
| | - Carla Barbosa Nonino
- Laboratory of Nutrigenomic Studies, Ribeirão Preto Medical School, University of Sao Paulo (USP), Ribeirao Preto, SP, Brazil
| | - Rebeca A Beraldo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo (USP), Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirao Preto, SP, 14049-900, Brazil
| | - Maria Cristina Foss-Freitas
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo (USP), Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirao Preto, SP, 14049-900, Brazil.
| |
Collapse
|
17
|
Qiu J, Zhang Z, Wang S, Chen Y, Liu C, Xu S, Wang D, Su J, Ni M, Yu J, Cui X, Ma L, Hu T, Hu Y, Gu X, Ma X, Wang J, Xu L. Transferrin Receptor Functionally Marks Thermogenic Adipocytes. Front Cell Dev Biol 2020; 8:572459. [PMID: 33251209 PMCID: PMC7676909 DOI: 10.3389/fcell.2020.572459] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
Background Thermogenic adipocytes, including beige and brown adipocytes, are critical for thermogenesis and energy homeostasis. Identification of functional cell surface markers of thermogenic adipocytes is of significance for potential application in biological and clinical practices. Methods With a combination of RNA-sequencing of in vivo and in vitro models, we identified transferrin receptor (Tfr1), a receptor specialized for cellular iron uptake, as a previously unappreciated cell surface molecule for thermogenic adipocytes compared to white adipocytes. The alternation of Tfr1 levels under physiological and pathological stimuli was assessed, and the mitochondria functionality, browning capacity, and iron metabolism of mature adipocytes were examined with Tfr1 knockdown. Results Tfr1 was expressed predominantly in thermogenic adipocytes versus white adipocyte, and its expression levels were tightly correlated with the activation or inhibition status of thermogenic adipocytes under external stimuli. Besides, Tfr1 gene deficiency in thermogenic adipocytes led to reduced thermogenic gene programs and mitochondrial integrity. Conclusion Tfr1 functionally marks thermogenic adipocytes and could serve as a potential thermogenic adipocyte surface marker.
Collapse
Affiliation(s)
- Jin Qiu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Zhiyin Zhang
- Department of Endocrinology and Metabolism, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sainan Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yanru Chen
- Department of Endocrinology and Metabolism, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Caizhi Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Sainan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Dongmei Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Junlei Su
- Department of Endocrinology and Metabolism, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengshan Ni
- Department of Endocrinology and Metabolism, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Yu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Xiangdi Cui
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Lu Ma
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Tianhui Hu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yepeng Hu
- Department of Endocrine and Metabolic Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xuejiang Gu
- Department of Endocrine and Metabolic Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinran Ma
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China.,Department of Endocrine and Metabolic Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiqiu Wang
- Department of Endocrinology and Metabolism, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
18
|
Souza de Oliveira M, Sachs Nique P, Crispim D, Marmontel de Souza B. The association of uncoupling proteins 1, 2, and 3 with weight loss variability after bariatric surgery: a systematic review. Surg Obes Relat Dis 2020; 16:1858-1868. [DOI: 10.1016/j.soard.2020.06.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/05/2020] [Accepted: 06/11/2020] [Indexed: 12/12/2022]
|
19
|
Ilyas S, Al-Refai R, Maharjan R, Diaz Bustamante L, Ghattas KN, Khan S. Bariatric Surgery and Type 2 Diabetes Mellitus: Assessing Factors Leading to Remission. A Systematic Review. Cureus 2020; 12:e9973. [PMID: 32983676 PMCID: PMC7510520 DOI: 10.7759/cureus.9973] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 08/23/2020] [Indexed: 12/05/2022] Open
Abstract
Type 2 Diabetes Mellitus (T2DM) is a health problem of paramount proportions and is associated with significant morbidity and mortality. Our study aims to review data published on the effects of different types of bariatric surgeries on T2DM remission, compared to lifestyle and medical intervention (LMI) exclusively, along with a comprehensive finding of numerous preoperative factors that lead to remission. We used PubMed, PubMed Central (PMC), and MEDLINE to search for literature. Our criteria included peer-reviewed, English language articles published in 2010 and onwards, consisting of adults with T2DM and a body mass index (BMI) of >30 kg/m2 as the population of interest. Twenty-four articles with 5,411 patients were selected for this systematic review, which included nine randomized controlled trials (RCTs) and 15 observational studies. The primary endpoint was T2DM remission. Based on the review, bariatric surgery is superior to LMI in inducing remission in T2DM, especially when employing the Roux-en-Y Gastric Bypass (RYGB) technique. Lower age of onset and shorter duration of T2DM, along with a high BMI are some of the factors that can lead to greater remission rates. Further research in RCTs is needed by incorporating double/triple-blind protocols, a standard definition of T2DM remission, long follow-up periods to evaluate for relapses in remission and any side effects, with a focus on inflammatory markers (eg, osteopontin), scoring systems (eg, DiaRem), and benefits of One-Anastomosis Gastric Bypass (OAGB) over other modalities, to advance our understanding of T2DM remission.
Collapse
Affiliation(s)
- Shahbakht Ilyas
- Surgery, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
- Medicine and Surgery, CMH Lahore Medical College and Institute of Dentistry, Lahore, PAK
| | - Reham Al-Refai
- Pathology, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Reeju Maharjan
- Neurology, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Liliana Diaz Bustamante
- Family Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Kyrillos N Ghattas
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
- Internal Medicine, Assiut University, Assiut, EGY
| | - Safeera Khan
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| |
Collapse
|
20
|
Gordaliza‐Alaguero I, Cantó C, Zorzano A. Metabolic implications of organelle-mitochondria communication. EMBO Rep 2019; 20:e47928. [PMID: 31418169 PMCID: PMC6726909 DOI: 10.15252/embr.201947928] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/10/2019] [Accepted: 05/28/2019] [Indexed: 12/31/2022] Open
Abstract
Cellular organelles are not static but show dynamism-a property that is likely relevant for their function. In addition, they interact with other organelles in a highly dynamic manner. In this review, we analyze the proteins involved in the interaction between mitochondria and other cellular organelles, especially the endoplasmic reticulum, lipid droplets, and lysosomes. Recent results indicate that, on one hand, metabolic alterations perturb the interaction between mitochondria and other organelles, and, on the other hand, that deficiency in proteins involved in the tethering between mitochondria and the ER or in specific functions of the interaction leads to metabolic alterations in a variety of tissues. The interaction between organelles is an emerging field that will permit to identify key proteins, to delineate novel modulation pathways, and to elucidate their implications in human disease.
Collapse
Affiliation(s)
- Isabel Gordaliza‐Alaguero
- Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
- CIBER de Diabetes y Enfermedades Metabolicas AsociadasBarcelonaSpain
- Departamento de Bioquimica i Biomedicina MolecularFacultat de BiologiaUniversitat de BarcelonaBarcelonaSpain
| | - Carlos Cantó
- Nestle Institute of Health Sciences (NIHS)LausanneSwitzerland
- School of Life SciencesEcole Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
- CIBER de Diabetes y Enfermedades Metabolicas AsociadasBarcelonaSpain
- Departamento de Bioquimica i Biomedicina MolecularFacultat de BiologiaUniversitat de BarcelonaBarcelonaSpain
| |
Collapse
|
21
|
Courcoulas AP, Stefater MA, Shirley E, Gourash WF, Stylopoulos N. The Feasibility of Examining the Effects of Gastric Bypass Surgery on Intestinal Metabolism: Prospective, Longitudinal Mechanistic Clinical Trial. JMIR Res Protoc 2019; 8:e12459. [PMID: 30679147 PMCID: PMC6483060 DOI: 10.2196/12459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/05/2018] [Accepted: 12/09/2018] [Indexed: 01/01/2023] Open
Abstract
Background Bariatric surgery, especially Roux-en-Y gastric bypass (RYGB), is the best treatment for severe obesity and its complications including type 2 diabetes mellitus (T2DM). Understanding the mechanisms responsible for the beneficial metabolic effects will help to engineer ways to improve the procedure or produce these effects without surgery. Objective The aim is to present data on recruitment and feasibility of a translational study designed to collect intestinal samples before and after bariatric surgery. The goal of biobanking is to allow future studies to test the hypothesis that the mechanism of action of RYGB involves specific changes in the postsurgical short- and long-term metabolism and morphology of the jejunum (Roux limb). Specifically, to test whether the intestine enhances its metabolism and activity after RYGB and increases its fuel utilization, we designed a prospective, longitudinal study, which involved the recruitment of candidates for RYGB with and without T2DM. We describe the tissue bank that we have generated, and our experience, hoping to further facilitate the performance of longitudinal mechanistic studies in human patients undergoing bariatric surgery and especially those involving post-RYGB intestinal biology. Methods We conducted a trial to characterize the effects of RYGB on intestinal metabolism. Intestinal tissue samples were collected from the jejunum at surgery, 1, 6, and 12 months postoperatively for the analysis of intestinal gene expression and metabolomic and morphologic changes. The target number of patients who completed at least the 6-month follow-up was 26, and we included a 20% attrition rate, increasing the total number to 32. Results To enroll 26 patients, we had to approach 79 potential participants. A total of 37 agreed to participate and started the study; 33, 30, and 26 active participants completed their 1-month, 6-month, and 12-month studies, respectively. Three participants withdrew, and 30 participants are still active. Altruism and interest in research were the most common reasons for participation. Important factors for feasibility and successful retention included (1) large volume case flow, (2) inclusion and exclusion criteria broad enough to capture a large segment of the patient population but narrow enough to ensure the completion of study aims and protection of safety concerns, (3) accurate assessment of willingness and motivation to participate in a study, (4) seamless integration of the recruitment process into normal clinical flow, (5) financial reimbursement and nonfinancial rewards and gestures of appreciation, and (6) nonburdensome follow-up visits and measures and reasonable time allotted. Conclusions Human translational studies of the intestinal mechanisms of metabolic and weight changes after bariatric surgery are important and feasible. A tissue bank with unique samples has been established that could be used by investigators in many research fields, further enabling mechanistic studies on the effects of bariatric surgery. Trial Registration ClinicalTrials.gov NCT02710370; https://clinicaltrials.gov/ct2/show/NCT02710370 (Archived by WebCite at http://www.webcitation.org/75HrQT8Dl)
Collapse
Affiliation(s)
| | | | - Eleanor Shirley
- University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - William F Gourash
- University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | | |
Collapse
|
22
|
Kristensen MD, Petersen SM, Møller KE, Lund MT, Hansen M, Hansen CN, Courraud J, Helge JW, Dela F, Prats C. Obesity leads to impairments in the morphology and organization of human skeletal muscle lipid droplets and mitochondrial networks, which are resolved with gastric bypass surgery-induced improvements in insulin sensitivity. Acta Physiol (Oxf) 2018; 224:e13100. [PMID: 29791782 DOI: 10.1111/apha.13100] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 12/29/2022]
Abstract
AIMS Skeletal muscle lipid stores and mitochondrial function have been appointed as key players in obesity-induced insulin resistance. However, there are conflicting reports in the literature based on in vitro quantitative measurements. Here, we test the hypothesis that it is not the quantity but the quality that matters. METHODS This study combines quantitative and qualitative structural measurements of lipid stores and mitochondrial dynamics in skeletal muscle from lean subjects, and subjects with morbid obesity, with and without type 2 diabetes, before and after gastric bypass surgery. RESULTS The structural organization of muscle mitochondrial networks in type II muscle fibres from subjects with morbid obesity is impaired. In addition, the amount of skeletal muscle perilipin 2 protein per intramyocellular lipid is reduced in subjects with morbid obesity, resulting in qualitative alterations in perilipin 2 coat around some lipid droplets. Gastric bypass surgery-induced weight loss and insulin resistance remission were associated with decreases in intramyocellular lipid stores and, qualitative improvements in lipid droplets' morphology, perilipin 2 coat and mitochondrial dynamics. CONCLUSION Morbid obesity leads to severe qualitative alterations of both skeletal muscle lipid stores and mitochondrial networks. The degree of structural improvements after gastric bypass surgery was proportional to the improvements in whole body insulin sensitivity, suggesting an association between these events.
Collapse
Affiliation(s)
- M. D. Kristensen
- X-lab, Center for Healthy Aging; University of Copenhagen; Copenhagen Denmark
| | - S. M. Petersen
- X-lab, Center for Healthy Aging; University of Copenhagen; Copenhagen Denmark
| | - K. E. Møller
- X-lab, Center for Healthy Aging; University of Copenhagen; Copenhagen Denmark
| | - M. T. Lund
- X-lab, Center for Healthy Aging; University of Copenhagen; Copenhagen Denmark
| | - M. Hansen
- X-lab, Center for Healthy Aging; University of Copenhagen; Copenhagen Denmark
| | - C. N Hansen
- X-lab, Center for Healthy Aging; University of Copenhagen; Copenhagen Denmark
| | - J. Courraud
- X-lab, Center for Healthy Aging; University of Copenhagen; Copenhagen Denmark
| | - J. W. Helge
- X-lab, Center for Healthy Aging; University of Copenhagen; Copenhagen Denmark
| | - F. Dela
- X-lab, Center for Healthy Aging; University of Copenhagen; Copenhagen Denmark
- Department of Geriatrics; Bispebjerg University Hospital; Copenhagen Denmark
| | - C. Prats
- X-lab, Center for Healthy Aging; University of Copenhagen; Copenhagen Denmark
- Core Facility for Integrated Microscopy; Department of Biomedical Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| |
Collapse
|
23
|
Lu Z, Wei X, Sun F, Zhang H, Gao P, Pu Y, Wang A, Chen J, Tong W, Li Q, Zhou X, Yan Z, Zheng H, Yang G, Huang Y, Liu D, Zhu Z. Non-insulin determinant pathways maintain glucose homeostasis upon metabolic surgery. Cell Discov 2018; 4:58. [PMID: 30275974 PMCID: PMC6155125 DOI: 10.1038/s41421-018-0062-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/13/2018] [Accepted: 08/30/2018] [Indexed: 12/16/2022] Open
Abstract
Insulin is critical for glucose homeostasis, and insulin deficiency or resistance leads to the development of diabetes. Recent evidence suggests that diabetes can be remitted independent of insulin. However, the underlying mechanism remains largely elusive. In this study, we utilized metabolic surgery as a tool to identify the non-insulin determinant mechanism. Here, we report that the most common metabolic surgery, Roux-en-Y gastric bypass (RYGB), reduced insulin production but persistently maintained euglycemia in healthy Sprague-Dawley (SD) rats and C57 mice. This reduction in insulin production was associated with RYGB-mediated inhibition of pancreatic preproinsulin and polypyrimidine tract-binding protein 1. In addition, RYGB also weakened insulin sensitivity that was evaluated by hyperinsulinemic-euglycemic clamp test and downregulated signaling pathways in insulin-sensitive tissues. The mechanistic evidence suggests that RYGB predominately shifted the metabolic profile from glucose utilization to fatty acid oxidation, enhanced the energy expenditure and activated multiple metabolic pathways through reducing gut energy uptake. Importantly, the unique effect of RYGB was extended to rats with islet disruption and patients with type 2 diabetes. These results demonstrate that compulsory rearrangement of the gastrointestinal tract can initiate non-insulin determinant pathways to maintain glucose homeostasis. Based on the principle of RYGB action, the development of a noninvasive intervention of the gastrointestinal tract is a promising therapeutic route to combat disorders characterized by energy metabolism dysregulation.
Collapse
Affiliation(s)
- Zongshi Lu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Xiao Wei
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Fang Sun
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Hexuan Zhang
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Peng Gao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Yunfei Pu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Anlong Wang
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Jing Chen
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Weidong Tong
- Department of Gastrointestinal Metabolic Surgery, Daping Hospital, Third Military Medical University, Chongqing, 400042 China
| | - Qiang Li
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Xunmei Zhou
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Zhencheng Yan
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Hongting Zheng
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037 China
| | - Gangyi Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010 China
| | - Yu Huang
- Institute of Vascular Medicine and School of Biomedical Sciences, Chinese University of Hong Kong, BMSB315, Shatin, Hong Kong 00852 China
| | - Daoyan Liu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400042 China
| |
Collapse
|
24
|
Balampanis K, Chasapi A, Kourea E, Tanoglidi A, Hatziagelaki E, Lambadiari V, Dimitriadis G, Lambrou GI, Kalfarentzos F, Melachrinou M, Sotiropoulou-Bonikou G. Inter-tissue expression patterns of the key metabolic biomarker PGC-1α in severely obese individuals: Implication in obesity-induced disease. Hellenic J Cardiol 2018; 60:282-293. [PMID: 30138744 DOI: 10.1016/j.hjc.2018.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/29/2018] [Accepted: 08/03/2018] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE PGC-1α is already known as a significant regulator of mitochondrial biogenesis, oxidative phosphorylation and fatty acid metabolism. Our study focuses on the role of PGC1α in morbid obesity, in five different tissues, collected from 50 severely obese patients during planned bariatric surgery. METHODS The investigated tissues included subcutaneous adipose tissue (SAT), visceral adipose tissue (VAT), skeletal muscle (SM), extramyocellular adipose tissue (EMAT) and liver. PGC1α expression was investigated with immunohistochemistry and evaluated with microscopy. RESULTS Our findings highlighted significant positive inter-tissue correlations regarding PGC-1α expression between several tissue pairs (VAT-SAT, VAT-SM, VAT-EMAT, SAT-SM, SAT-EMAT, SM-EMAT). Moreover, we found significant negative correlations between PGC1α expression in VAT with CD68 expression in skeletal muscle and EMAT, implying a possible protective role of PGC1α against obesity-induced inflammation. CONCLUSION Unmasking the inter-tissue communication networks regarding PGC-1α expression in morbid obesity, will give more insight into its significant role in obesity-induced diseases. PGC1α could potentially represent a future preventive and therapeutic target against obesity-induced disease, probably through enhancing mitochondrial biogenesis and metabolism.
Collapse
Affiliation(s)
- Konstantinos Balampanis
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece; Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece.
| | - Athina Chasapi
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece.
| | - Eleni Kourea
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece.
| | - Anna Tanoglidi
- Department of Clinical Pathology, Akademiska University, Uppsala, Sweden.
| | - Erifili Hatziagelaki
- Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece.
| | - Vaia Lambadiari
- Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece.
| | - George Dimitriadis
- Second Department of Internal Medicine, Research Unit and Diabetes Center, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Rimini 1, Haidari, 12462 Athens, Greece.
| | - George I Lambrou
- First Department of Pediatrics, Choremeio Research Laboratory, National and Kapodistrian University of Athens, Medical School, Thivon & Levadeias 8, Goudi, 11527 Athens, Greece.
| | - Fotios Kalfarentzos
- Department of Surgery, Medical School, University of Patras, 26500 Patras, Greece.
| | - Maria Melachrinou
- Department of Pathology, Medical School, University of Patras, 26500 Patras, Greece.
| | | |
Collapse
|
25
|
Sacks J, Mulya A, Fealy CE, Huang H, Mosinski JD, Pagadala MR, Shimizu H, Batayyah E, Schauer PR, Brethauer SA, Kirwan JP. Effect of Roux-en-Y gastric bypass on liver mitochondrial dynamics in a rat model of obesity. Physiol Rep 2018; 6:e13600. [PMID: 29464885 PMCID: PMC5820430 DOI: 10.14814/phy2.13600] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 12/22/2017] [Accepted: 01/09/2018] [Indexed: 12/15/2022] Open
Abstract
Bariatric surgery provides significant and durable improvements in glycemic control and hepatic steatosis, but the underlying mechanisms that drive improvements in these metabolic parameters remain to be fully elucidated. Recently, alterations in mitochondrial morphology have shown a direct link to nutrient adaptations in obesity. Here, we evaluate the effects of Roux-en-Y gastric bypass (RYGB) surgery on markers of liver mitochondrial dynamics in a diet-induced obesity Sprague-Dawley (SD) rat model. Livers were harvested from adult male SD rats 90-days after either Sham or RYGB surgery and continuous high-fat feeding. We assessed expression of mitochondrial proteins involved in fusion, fission, mitochondrial autophagy (mitophagy) and biogenesis, as well as differences in citrate synthase activity and markers of oxidative stress. Gene expression for mitochondrial fusion genes, mitofusin 1 (Mfn1; P < 0.05), mitofusin 2 (Mfn2; P < 0.01), and optic atrophy 1 (OPA1; P < 0.05) increased following RYGB surgery. Biogenesis regulators, peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α; P < 0.01) and nuclear respiratory factor 1 (Nrf1; P < 0.05), also increased in the RYGB group, as well as mitophagy marker, BCL-2 interacting protein 3 (Bnip3; P < 0.01). Protein expression for Mfn1 (P < 0.001), PGC1α (P < 0.05), BNIP3 (P < 0.0001), and mitochondrial complexes I-V (P < 0.01) was also increased by RYGB, and Mfn1 expression negatively correlated with body weight, insulin resistance, and fasting plasma insulin. In the RYGB group, citrate synthase activity was increased (P < 0.02) and reactive oxygen species (ROS) was decreased compared to the Sham control group (P < 0.05), although total antioxidant capacity was unchanged between groups. These data are the first to show an association between RYGB surgery and improved markers of liver mitochondrial dynamics. These observed improvements may be related to weight loss and reduced energetic demand on the liver, which could facilitate normalization of glucose homeostasis and protect against hepatic steatosis.
Collapse
Affiliation(s)
- Jessica Sacks
- Department of PathobiologyLerner Research InstituteCleveland ClinicClevelandOhio
- Molecular MedicineCleveland Clinic Lerner College of MedicineCase Western Reserve UniversityClevelandOhio
| | - Anny Mulya
- Department of PathobiologyLerner Research InstituteCleveland ClinicClevelandOhio
| | - Ciaran E. Fealy
- Department of PathobiologyLerner Research InstituteCleveland ClinicClevelandOhio
| | - Hazel Huang
- Department of PathobiologyLerner Research InstituteCleveland ClinicClevelandOhio
| | - John D. Mosinski
- Department of PathobiologyLerner Research InstituteCleveland ClinicClevelandOhio
| | - Mangesh R. Pagadala
- Department of Gastroenterology and HepatologyDigestive Disease InstituteCleveland ClinicClevelandOhio
| | | | - Esam Batayyah
- Bariatric and Metabolic InstituteCleveland ClinicClevelandOhio
| | - Philip R. Schauer
- Bariatric and Metabolic InstituteCleveland ClinicClevelandOhio
- Metabolic Translational Research CenterEndocrinology and Metabolism InstituteCleveland ClinicClevelandOhio
| | - Stacy A. Brethauer
- Bariatric and Metabolic InstituteCleveland ClinicClevelandOhio
- Metabolic Translational Research CenterEndocrinology and Metabolism InstituteCleveland ClinicClevelandOhio
| | - John P. Kirwan
- Department of PathobiologyLerner Research InstituteCleveland ClinicClevelandOhio
- Department of Gastroenterology and HepatologyDigestive Disease InstituteCleveland ClinicClevelandOhio
- Bariatric and Metabolic InstituteCleveland ClinicClevelandOhio
- Metabolic Translational Research CenterEndocrinology and Metabolism InstituteCleveland ClinicClevelandOhio
| |
Collapse
|
26
|
Jiang Z, Knudsen NH, Wang G, Qiu W, Naing ZZC, Bai Y, Ai X, Lee CH, Zhou X. Genetic Control of Fatty Acid β-Oxidation in Chronic Obstructive Pulmonary Disease. Am J Respir Cell Mol Biol 2017; 56:738-748. [PMID: 28199134 PMCID: PMC5516290 DOI: 10.1165/rcmb.2016-0282oc] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 01/31/2017] [Indexed: 12/17/2022] Open
Abstract
Bioenergetics homeostasis is important for cells to sustain normal functions and defend against injury. The genetic controls of bioenergetics homeostasis, especially lipid metabolism, remain poorly understood in chronic obstructive pulmonary disease (COPD), the third leading cause of death in the world. Additionally, the biological function of most of the susceptibility genes identified from genome-wide association studies (GWASs) in COPD remains unclear. Here, we aimed to address (1) how fatty acid oxidation (FAO), specifically β-oxidation, a key lipid metabolism pathway that provides energy to cells, contributes to cigarette smoke (CS)-induced COPD; and (2) whether-and if so, how-FAM13A (family with sequence similarity 13 member A), a well-replicated COPD GWAS gene, modulates the FAO pathway. We demonstrated that CS induced expression of carnitine palmitoyltransferase 1A (CPT1A), a key mitochondrial enzyme for the FAO pathway, thereby enhancing FAO. Pharmacological inhibition of FAO by etomoxir blunted CS-induced reactive oxygen species accumulation and cell death in lung epithelial cells. FAM13A promoted FAO, possibly by interacting with and activating sirutin 1, and increasing expression of CPT1A. Furthermore, CS-induced cell death was reduced in lungs from Fam13a-/- mice. Our results suggest that FAM13A, the COPD GWAS gene, shapes the cellular metabolic response to CS exposure by promoting the FAO pathway, which may contribute to COPD development.
Collapse
Affiliation(s)
| | - Nelson H. Knudsen
- Departments of Genetics and Complex Diseases, and
- Nutrition, Division of Biological Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, and
| | - Gang Wang
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts
| | | | | | - Yan Bai
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Xingbin Ai
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Chih-Hao Lee
- Departments of Genetics and Complex Diseases, and
- Nutrition, Division of Biological Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, and
| | - Xiaobo Zhou
- Channing Division of Network Medicine and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| |
Collapse
|
27
|
Boulinguiez A, Staels B, Duez H, Lancel S. Mitochondria and endoplasmic reticulum: Targets for a better insulin sensitivity in skeletal muscle? Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:901-916. [PMID: 28529179 DOI: 10.1016/j.bbalip.2017.05.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 05/15/2017] [Accepted: 05/17/2017] [Indexed: 12/16/2022]
Abstract
Obesity and its associated metabolic disorders represent a major health burden, with economic and social consequences. Although adapted lifestyle and bariatric surgery are effective in reducing body weight, obesity prevalence is still rising. Obese individuals often become insulin-resistant. Obesity impacts on insulin responsive organs, such as the liver, adipose tissue and skeletal muscle, and increases the risk of cardiovascular diseases, type 2 diabetes and cancer. In this review, we discuss the effects of obesity and insulin resistance on skeletal muscle, an important organ for the control of postprandial glucose. The roles of mitochondria and the endoplasmic reticulum in insulin signaling are highlighted and potential innovative research and treatment perspectives are proposed.
Collapse
Affiliation(s)
- Alexis Boulinguiez
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000, Lille, France.
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000, Lille, France.
| | - Hélène Duez
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000, Lille, France.
| | - Steve Lancel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000, Lille, France.
| |
Collapse
|
28
|
Pardina E, Ferrer R, Rossell J, Ricart-Jané D, Méndez-Lara KA, Baena-Fustegueras JA, Lecube A, Julve J, Peinado-Onsurbe J. Hepatic CD36 downregulation parallels steatosis improvement in morbidly obese undergoing bariatric surgery. Int J Obes (Lond) 2017; 41:1388-1393. [PMID: 28555086 DOI: 10.1038/ijo.2017.115] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 02/28/2017] [Accepted: 04/02/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND The notion that hepatic expression of genes involved in lipid metabolism is altered in obese patients is relatively new and its relationship with hepatic steatosis and cardiometabolic alterations remains unclear. OBJECTIVE We assessed the impact of Roux-en-Y gastric bypass surgery (RYGB) on the expression profile of genes related to metabolic syndrome in liver biopsies from morbidly obese individuals using a custom-made, focused cDNA microarray, and assessed the relationship between the expression profile and hepatic steatosis regression. MATERIALS AND METHODS Plasma and liver samples were obtained from patients at baseline and 12 months after surgery. Samples were assayed for chemical and gene expression analyses, as appropriate. Gene expression profiles were assessed using custom-made, focused TaqMan low-density array cards. RESULTS RYGB-induced weight loss produced a favorable reduction in fat deposits, insulin resistance (estimated by homeostasis model assessment of insulin resistance (HOMA-IR)), and plasma and hepatic lipid levels. Compared with the baseline values, the gene expression levels of key targets of lipid metabolism were significantly altered: CD36 was significantly downregulated (-40%; P=0.001), whereas APOB (+27%; P=0.032) and SCARB1 (+37%; P=0.040) were upregulated in response to surgery-induced weight reduction. We also observed a favorable reduction in the expression of the PAI1 gene (-80%; P=0.007) and a significant increase in the expression of the PPARA (+60%; P=0.014) and PPARGC1 genes (+36%; P=0.015). Notably, the relative fold decrease in the expression of the CD36 gene was directly associated with a concomitant reduction in the cholesterol (Spearman's r=0.92; P=0.001) and phospholipid (Spearman's r=0.76; P=0.04) contents in this tissue. CONCLUSIONS For the first time, RYGB-induced weight loss was shown to promote a favorable downregulation of CD36 expression, which was proportional to a favorable reduction in the hepatic cholesterol and phospholipid contents in our morbidly obese subjects following surgery.
Collapse
Affiliation(s)
- E Pardina
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - R Ferrer
- Unitat d'Hormones, Servei de Bioquímica, Hospital Universitari de la Vall d'Hebron, Barcelona, Spain
| | - J Rossell
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - D Ricart-Jané
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - K A Méndez-Lara
- Institut de Recerca de l'Hospital de La Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques de l'Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | | | - A Lecube
- Departament d'Endocrinologia i Nutrició, Hospital Universitari Arnau de Vilanova, Universitat de Lleida, Lleida, Spain.,Unitat de Recerca en Diabetes i Metabolisme, Institut de Recerca Hospital Universitari Vall d'Hebron, Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Barcelona, Spain
| | - J Julve
- Institut de Recerca de l'Hospital de La Santa Creu i Sant Pau, Institut d'Investigacions Biomèdiques de l'Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain.,Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Barcelona, Spain
| | - J Peinado-Onsurbe
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
29
|
Zhao L, Zhu L, Su Z, Liu Y, Li P, Yang X, Li W, Tan L, Sun X, Zhu S. Using the hyperinsulinemic euglycemic clamp to assess insulin sensitivity at 3 months following Roux-en-Y gastric bypass surgery in type 2 diabetes patients with BMI <35 kg/m 2 in China. Int J Surg 2016; 38:90-94. [PMID: 28043929 DOI: 10.1016/j.ijsu.2016.12.120] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 12/14/2016] [Accepted: 12/26/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND The aim of this study was to describe short-time effect of Roux-en-Y gastric bypass (RYGB) on insulin sensitivity (IS) of Chinese diabetes patients with body mass index (BMI) <35 kg/m2 by hyperinsulinemic euglycemic clamp. MATERIALS AND METHODS We studied 15 type 2 diabete mellitus (T2DM) patients with BMI <35 kg/m2, who underwent laparoscopical Roux-en-Y gastric bypass (LRYGB). Hyperinsulinemic-euglycemic clamp were performed at baseline and at 3 months after LRYGB. RESULTS The glucose disposal rate (M value) increased significantly at 3 months after RYGB (from 3.36 ± 1.26 mg kg-1 min-1 to 6.30 ± 1.3 mg kg-1 min-1, p < 0.001). The time to reach euglycemia at the hyperinsulinemic euglycemic clamp reduced remarkably from baseline to 3 months after RYGB (from 114.40 ± 6.11 min to 97.93 ± 8.57 min, p < 0.001). There was a marked reduction in value of HOMA-IR (from 4.47 ± 2.20 mg kg-1 min-1 to 2.10 ± 0.75 mg kg-1 min-1, p < 0.001). The parameters of body fat distribution (body weight, BMI, waist circumference, waist to hip radio) changed obviously after surgery. RYGB caused a significant improvement in fasting and postprandial plasma glucose and insulin, HbAc1. Preoperative M value and the time to reach steady-state correlated with changes of M and the time to reach steady-state at 3 months after RYGB. CONCLUSION Peripheral and hepatic IS improved remarkably at 3 months following RYGB, as an important mechanism for early improvement in T2DM patients with low BMI. And the time to reach euglycemia at the hyperinsulinemic euglycemic clamp may be an useful index of assessing insulin sensitivity. It is more reasonal to combine the time to reach euglycemia with M value for assessing IS.
Collapse
Affiliation(s)
- Lei Zhao
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan, People's Republic of China
| | - Liyong Zhu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan, People's Republic of China
| | - Zhihong Su
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan, People's Republic of China
| | - Yong Liu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan, People's Republic of China
| | - Pengzhou Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan, People's Republic of China
| | - Xiangwu Yang
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan, People's Republic of China
| | - Weizheng Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan, People's Republic of China
| | - Lingjie Tan
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan, People's Republic of China
| | - Xulong Sun
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan, People's Republic of China
| | - Shaihong Zhu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan, People's Republic of China.
| |
Collapse
|
30
|
Campbell LE, Langlais PR, Day SE, Coletta RL, Benjamin TR, De Filippis EA, Madura JA, Mandarino LJ, Roust LR, Coletta DK. Identification of Novel Changes in Human Skeletal Muscle Proteome After Roux-en-Y Gastric Bypass Surgery. Diabetes 2016; 65:2724-31. [PMID: 27207528 PMCID: PMC5001187 DOI: 10.2337/db16-0004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 04/29/2016] [Indexed: 12/18/2022]
Abstract
The mechanisms of metabolic improvements after Roux-en-Y gastric bypass (RYGB) surgery are not entirely clear. Therefore, the aim of our study was to investigate the role of obesity and RYGB on the human skeletal muscle proteome. Basal muscle biopsies were obtained from seven obese (BMI >40 kg/m(2)) female subjects (45.1 ± 3.6 years) pre- and 3 months post-RYGB, and euglycemic-hyperinsulinemic clamps were used to assess insulin sensitivity. Four age-matched (48.5 ± 4.7 years) lean (BMI <25 kg/m(2)) females served as control subjects. We performed quantitative mass spectrometry and microarray analyses on protein and RNA isolated from the muscle biopsies. Significant improvements in fasting plasma glucose (104.2 ± 7.8 vs. 86.7 ± 3.1 mg/dL) and BMI (42.1 ± 2.2 vs. 35.3 ± 1.8 kg/m(2)) were demonstrated in the pre- versus post-RYGB, both P < 0.05. Proteomic analysis identified 2,877 quantifiable proteins. Of these, 395 proteins were significantly altered in obesity before surgery, and 280 proteins differed significantly post-RYGB. Post-RYGB, 49 proteins were returned to normal levels after surgery. KEGG pathway analysis revealed a decreased abundance in ribosomal and oxidative phosphorylation proteins in obesity, and a normalization of ribosomal proteins post-RYGB. The transcriptomic data confirmed the normalization of the ribosomal proteins. Our results provide evidence that obesity and RYGB have a dynamic effect on the skeletal muscle proteome.
Collapse
Affiliation(s)
| | | | - Samantha E Day
- School of Life Sciences, Arizona State University, Tempe, AZ
| | - Richard L Coletta
- School for the Science of Health Care Delivery, Arizona State University, Phoenix, AZ
| | | | | | | | - Lawrence J Mandarino
- Mayo Clinic, Scottsdale, AZ School for the Science of Health Care Delivery, Arizona State University, Phoenix, AZ
| | | | - Dawn K Coletta
- Mayo Clinic, Scottsdale, AZ School for the Science of Health Care Delivery, Arizona State University, Phoenix, AZ Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ
| |
Collapse
|
31
|
Coen PM, Menshikova EV, Distefano G, Zheng D, Tanner CJ, Standley RA, Helbling NL, Dubis GS, Ritov VB, Xie H, Desimone ME, Smith SR, Stefanovic-Racic M, Toledo FGS, Houmard JA, Goodpaster BH. Exercise and Weight Loss Improve Muscle Mitochondrial Respiration, Lipid Partitioning, and Insulin Sensitivity After Gastric Bypass Surgery. Diabetes 2015; 64:3737-50. [PMID: 26293505 PMCID: PMC4613980 DOI: 10.2337/db15-0809] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 07/30/2015] [Indexed: 01/03/2023]
Abstract
Both Roux-en-Y gastric bypass (RYGB) surgery and exercise can improve insulin sensitivity in individuals with severe obesity. However, the impact of RYGB with or without exercise on skeletal muscle mitochondria, intramyocellular lipids, and insulin sensitivity index (SI) is unknown. We conducted a randomized exercise trial in patients (n = 101) who underwent RYGB surgery and completed either a 6-month moderate exercise (EX) or a health education control (CON) intervention. SI was determined by intravenous glucose tolerance test. Mitochondrial respiration and intramyocellular triglyceride, sphingolipid, and diacylglycerol content were measured in vastus lateralis biopsy specimens. We found that EX provided additional improvements in SI and that only EX improved cardiorespiratory fitness, mitochondrial respiration and enzyme activities, and cardiolipin profile with no change in mitochondrial content. Muscle triglycerides were reduced in type I fibers in CON, and sphingolipids decreased in both groups, with EX showing a further reduction in a number of ceramide species. In conclusion, exercise superimposed on bariatric surgery-induced weight loss enhances mitochondrial respiration, induces cardiolipin remodeling, reduces specific sphingolipids, and provides additional improvements in insulin sensitivity.
Collapse
Affiliation(s)
- Paul M Coen
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, FL Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA Department of Health and Physical Activity, University of Pittsburgh, Pittsburgh, PA Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL
| | - Elizabeth V Menshikova
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Giovanna Distefano
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, FL Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA Department of Physical Therapy, University of Pittsburgh, Pittsburgh, PA
| | - Donghai Zheng
- Department of Kinesiology, East Carolina University, Greenville, NC
| | - Charles J Tanner
- Department of Kinesiology, East Carolina University, Greenville, NC
| | - Robert A Standley
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, FL Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Nicole L Helbling
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Gabriel S Dubis
- Department of Kinesiology, East Carolina University, Greenville, NC
| | - Vladimir B Ritov
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Hui Xie
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, FL
| | - Marisa E Desimone
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Steven R Smith
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, FL Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL
| | - Maja Stefanovic-Racic
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Frederico G S Toledo
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Joseph A Houmard
- Department of Kinesiology, East Carolina University, Greenville, NC
| | - Bret H Goodpaster
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, FL Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, FL
| |
Collapse
|
32
|
Albers PH, Bojsen-Møller KN, Dirksen C, Serup AK, Kristensen DE, Frystyk J, Clausen TR, Kiens B, Richter EA, Madsbad S, Wojtaszewski JFP. Enhanced insulin signaling in human skeletal muscle and adipose tissue following gastric bypass surgery. Am J Physiol Regul Integr Comp Physiol 2015; 309:R510-24. [PMID: 26062634 DOI: 10.1152/ajpregu.00228.2014] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 06/01/2015] [Indexed: 12/12/2022]
Abstract
Roux-en-Y gastric bypass (RYGB) leads to increased peripheral insulin sensitivity. The aim of this study was to investigate the effect of RYGB on expression and regulation of proteins involved in regulation of peripheral glucose metabolism. Skeletal muscle and adipose tissue biopsies from glucose-tolerant and type 2 diabetic subjects at fasting and during a hyperinsulinemic-euglycemic clamp before as well as 1 wk and 3 and 12 mo after RYGB were analyzed for relevant insulin effector proteins/signaling components. Improvement in peripheral insulin sensitivity mainly occurred at 12 mo postsurgery when major weight loss was evident and occurred concomitantly with alterations in plasma adiponectin and in protein expression/signaling in peripheral tissues. In skeletal muscle, protein expression of GLUT4, phosphorylated levels of TBC1D4, as well as insulin-induced changes in phosphorylation of Akt and glycogen synthase activity were enhanced 12 mo postsurgery. In adipose tissue, protein expression of GLUT4, Akt2, TBC1D4, and acetyl-CoA carboxylase (ACC), phosphorylated levels of AMP-activated protein kinase and ACC, as well as insulin-induced changes in phosphorylation of Akt and TBC1D4, were enhanced 12 mo postsurgery. Adipose tissue from glucose-tolerant subjects was the most responsive to RYGB compared with type 2 diabetic patients, whereas changes in skeletal muscle were largely similar in these two groups. In conclusion, an improved molecular insulin-sensitive phenotype of skeletal muscle and adipose tissue appears to contribute to the improved whole body insulin action following a substantial weight loss after RYGB.
Collapse
Affiliation(s)
- Peter H Albers
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, the August Krogh Centre, University of Copenhagen, Copenhagen, Denmark; Diabetes Research Unit, Novo Nordisk A/S, Maaloev, Denmark
| | - Kirstine N Bojsen-Møller
- Department of Endocrinology, Hvidovre Hospital, Hvidovre, Denmark; Novo Nordisk Foundation Centre for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; and
| | - Carsten Dirksen
- Department of Endocrinology, Hvidovre Hospital, Hvidovre, Denmark; Novo Nordisk Foundation Centre for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; and
| | - Annette K Serup
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, the August Krogh Centre, University of Copenhagen, Copenhagen, Denmark
| | - Dorte E Kristensen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, the August Krogh Centre, University of Copenhagen, Copenhagen, Denmark
| | - Jan Frystyk
- Medical Research Laboratory, Department of Clinical Medicine, Health, Aarhus University, Aarhus, Denmark
| | | | - Bente Kiens
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, the August Krogh Centre, University of Copenhagen, Copenhagen, Denmark
| | - Erik A Richter
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, the August Krogh Centre, University of Copenhagen, Copenhagen, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Hvidovre Hospital, Hvidovre, Denmark
| | - Jørgen F P Wojtaszewski
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, the August Krogh Centre, University of Copenhagen, Copenhagen, Denmark;
| |
Collapse
|
33
|
Zorzano A, Hernández-Alvarez MI, Sebastián D, Muñoz JP. Mitofusin 2 as a driver that controls energy metabolism and insulin signaling. Antioxid Redox Signal 2015; 22:1020-31. [PMID: 25567790 DOI: 10.1089/ars.2014.6208] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Mitochondrial dynamics is a complex process that impacts on mitochondrial biology. RECENT ADVANCES Recent evidence indicates that proteins participating in mitochondrial dynamics have additional cellular roles. Mitofusin 2 (Mfn2) is a potent modulator of mitochondrial metabolism with an impact on energy metabolism in muscle, liver, and hypothalamic neurons. In addition, Mfn2 is subjected to tight regulation. Hence, factors such as proinflammatory cytokines, lipid availability, or glucocorticoids block its expression, whereas exercise and increased energy expenditure promote its upregulation. CRITICAL ISSUES Importantly, Mfn2 controls cell metabolism and insulin signaling by limiting reactive oxygen species production and by modulation of endoplasmic reticulum stress. In this connection, it is critical to understand precisely the molecular mechanisms involved in the global actions of Mfn2. FUTURE DIRECTIONS Future directions should concentrate into the analysis of those mechanisms, and to fully demonstrate that Mfn2 represents a cellular hub that senses the metabolic and hormonal milieu and drives the control of metabolic homeostasis.
Collapse
Affiliation(s)
- Antonio Zorzano
- 1 Institute for Research in Biomedicine (IRB Barcelona) , Barcelona, Spain
| | | | | | | |
Collapse
|
34
|
Rieusset J. Contribution of mitochondria and endoplasmic reticulum dysfunction in insulin resistance: Distinct or interrelated roles? DIABETES & METABOLISM 2015; 41:358-68. [PMID: 25797073 DOI: 10.1016/j.diabet.2015.02.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 01/07/2015] [Accepted: 02/01/2015] [Indexed: 12/31/2022]
Abstract
Mitochondria and the endoplasmic reticulum (ER) regulate numerous cellular processes, and are critical contributors to cellular and whole-body homoeostasis. More important, mitochondrial dysfunction and ER stress are both closely associated with hepatic and skeletal muscle insulin resistance, thereby playing crucial roles in altered glucose homoeostasis in type 2 diabetes mellitus (T2DM). The accumulated evidence also suggests a potential interrelationship between alterations in both types of organelles, as mitochondrial dysfunction could participate in activation of the unfolded protein response, whereas ER stress could influence mitochondrial function. The fact that mitochondria and the ER are physically and functionally interconnected via mitochondria-associated membranes (MAMs) supports their interrelated roles in the pathophysiology of T2DM. However, the mechanisms that coordinate the interplay between mitochondrial dysfunction and ER stress, and its relevance to the control of glucose homoeostasis, are still unknown. This review evaluates the involvement of mitochondria and ER independently in the development of peripheral insulin resistance, as well as their potential roles in the disruption of organelle crosstalk at MAM interfaces in the alteration of insulin signalling.
Collapse
Affiliation(s)
- J Rieusset
- INSERM UMR-1060, CarMeN Laboratory, Lyon 1 University, INRA U1235, INSA of Lyon, Rockefeller and Charles-Merieux Lyon-Sud medical Universities, 69003 Lyon, France; Endocrinology, diabetology and nutrition service, Lyon-Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France.
| |
Collapse
|
35
|
Increased pyruvate dehydrogenase kinase expression in cultured myotubes from obese and diabetic individuals. Eur J Nutr 2014; 54:1033-43. [DOI: 10.1007/s00394-014-0780-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 10/06/2014] [Indexed: 12/11/2022]
|
36
|
He B, Piao D, Yu C, Wang Y, Han P. Amelioration in hepatic insulin sensitivity by reduced hepatic lipid accumulation at short-term after Roux-en-Y gastric bypass surgery in type 2 diabetic rats. Obes Surg 2014; 23:2033-41. [PMID: 23702909 DOI: 10.1007/s11695-013-0997-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Previous studies showed that early after Roux-en-Y gastric bypass (RYGB), there is a remarkable improvement in type 2 diabetes, which is characterized by insulin resistance. This study aims to gain insight into the underlying mechanisms of this effect. We determined the acute effects of RYGB on hepatic and peripheral insulin sensitivity. METHODS A rat model of type 2 diabetes was established using high-fat diet combined with streptozotocin (30 mg/kg, ip). Animals were divided into four groups: diabetic, diabetic RYGB, diabetic RYGB sham, and control rats. Hyperinsulinemic-euglycemic clamps with tracer infusion were completed at 2 weeks postoperatively to assess insulin sensitivity. Triglyceride concentration in liver and muscle tissues was determined. Protein kinase C (PKC) membrane translocation, protein expression of phospho-c-Jun NH2-terminal kinase (JNK), and phospho-IκB kinase β (IKKβ) were assessed with western blot. Malondialdehyde (MDA) and superoxide dismutase (SOD) activities in the liver were also measured. RESULTS RYGB surgery significantly improved hepatic insulin sensitivity index and decreased hepatic triglyceride concentration (P < 0.05), without an improvement in peripheral insulin sensitivity. Membrane translocation of PKC-ε, PKC-δ, and PKC-θ; the ratio of MDA to SOD; and the expression of p-JNK and p-IKKβ in the liver were lower in the diabetic RYGB group than in the diabetic group. CONCLUSIONS Diabetes remission was induced at short term after RYGB. The improvement of hepatic tissue lipotoxicity decreased the activation of certain PKC isoforms, the activity of JNK and IKK inflammatory signaling pathways, and the degree of oxidative stress. Furthermore, the hepatic insulin sensitivity was ameliorated, which is possibly a mechanism for early diabetes remission.
Collapse
Affiliation(s)
- Bing He
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, China,
| | | | | | | | | |
Collapse
|
37
|
Liu W, Beck BH, Vaidya KS, Nash KT, Feeley KP, Ballinger SW, Pounds KM, Denning WL, Diers AR, Landar A, Dhar A, Iwakuma T, Welch DR. Metastasis suppressor KISS1 seems to reverse the Warburg effect by enhancing mitochondrial biogenesis. Cancer Res 2013; 74:954-63. [PMID: 24351292 DOI: 10.1158/0008-5472.can-13-1183] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cancer cells tend to utilize aerobic glycolysis even under normoxic conditions, commonly called the "Warburg effect." Aerobic glycolysis often directly correlates with malignancy, but its purpose, if any, in metastasis remains unclear. When wild-type KISS1 metastasis suppressor is expressed, aerobic glycolysis decreases and oxidative phosphorylation predominates. However, when KISS1 is missing the secretion signal peptide (ΔSS), invasion and metastasis are no longer suppressed and cells continue to metabolize using aerobic glycolysis. KISS1-expressing cells have 30% to 50% more mitochondrial mass than ΔSS-expressing cells, which are accompanied by correspondingly increased mitochondrial gene expression and higher expression of PGC1α, a master coactivator that regulates mitochondrial mass and metabolism. PGC1α-mediated downstream pathways (i.e., fatty acid synthesis and β-oxidation) are differentially regulated by KISS1, apparently reliant upon direct KISS1 interaction with NRF1, a major transcription factor involved in mitochondrial biogenesis. Since the downstream effects could be reversed using short hairpin RNA to KISS1 or PGC1α, these data appear to directly connect changes in mitochondria mass, cellular glucose metabolism, and metastasis.
Collapse
Affiliation(s)
- Wen Liu
- Authors' Affiliations: Department of Cancer Biology; The University of Kansas Cancer Center, The University of Kansas Medical Center, Kansas City, Kansas; and Department of Pathology, University of Alabama-Birmingham, Birmingham, Alabama
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Longitudinal assessment of food intake, fecal energy loss, and energy expenditure after Roux-en-Y gastric bypass surgery in high-fat-fed obese rats. Obes Surg 2013; 23:531-40. [PMID: 23269513 DOI: 10.1007/s11695-012-0846-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND The efficacy of Roux-en-Y gastric bypass (RYGB) surgery to produce weight loss has been well-documented, but few studies have measured the key components of energy balance, food intake, and energy expenditure longitudinally. METHODS Male Sprague-Dawley rats on a high-fat diet underwent either RYGB, sham operation, or pair feeding and were compared to chow-fed lean controls. Body weight and composition, food intake and preference, energy expenditure, fecal output, and gastric emptying were monitored before and up to 4 months after intervention. RESULTS Despite the recovery of initially decreased food intake to levels slightly higher than before surgery and comparable to sham-operated rats after about 1 month, RYGB rats maintained a lower level of body weight and fat mass for 4 months that was not different from chow-fed age-matched controls. Energy expenditure corrected for lean body mass at 1 and 4 months after RYGB was not different from presurgical levels and from all other groups. Fecal energy loss was significantly increased at 6 and 16 weeks after RYGB compared to sham operation, and there was a progressive decrease in fat preference after RYGB. CONCLUSIONS In this rat model of RYGB, sustained weight loss is achieved by a combination of initial hypophagia and sustained increases in fecal energy loss, without change in energy expenditure per lean mass. A shift away from high-fat towards low-fat/high-carbohydrate food preference occurring in parallel suggests long-term adaptive mechanisms related to fat absorption.
Collapse
|
39
|
Pezeshki A, Chelikani PK. Effects of Roux-en-Y gastric bypass and ileal transposition surgeries on glucose and lipid metabolism in skeletal muscle and liver. Surg Obes Relat Dis 2013; 10:217-28. [PMID: 24603111 DOI: 10.1016/j.soard.2013.09.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 08/29/2013] [Accepted: 09/27/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND Roux-en Y gastric bypass (RYGB) and ileal transposition (IT) surgeries produce weight loss and improve diabetic control; however, the mechanisms of glycemic improvements are largely unknown. Because skeletal muscle and liver play a key role in glucose homeostasis, we compared the effects of RYGB and IT surgeries on key molecules of glucose and lipid metabolism in muscle and liver. METHODS Sprague-Dawley rats were subjected to RYGB, IT, or sham surgeries; sham-animals were ad-lib fed or pair-fed to RYGB rats (n = 7-9/group). At 8 weeks postoperatively, blood samples were collected for glucagon-like peptide-1 (GLP-1) and insulin analyses by ELISA. Leg muscle and liver tissues were analyzed for mRNA (RT-qPCR) and/or protein abundance (immuno blotting) of important molecules of glucose and lipid metabolism [glucose transporter-4 (GLUT-4), hexokinase, phosphofructokinase (PFK), adenosine monophosphate activated protein kinase-α (AMPKα), cytochrome C oxidase-IV (COX-IV), citrate synthase, carnitine palmitoyl transferase-1 (CPT-1), medium-chain acyl-CoA dehydrogenase (MCAD), peroxisome proliferator-activated receptor gamma co-activator 1 α (PGC-1 α), PGC-1-related coactivator (PRC), uncoupling protein-3 (UCP-3)]. RESULTS Plasma GLP-1 concentrations were increased comparably with RYGB and IT. RYGB and IT increased muscle GLUT-4 protein content, muscle hexokinase mRNA, and liver PFK mRNA. IT increased muscle AMPKα and COX-IV protein content and liver citrate synthase activity. IT increased muscle CPT-1, MCAD and PRC mRNA, whereas RYGB increased UCP-3 mRNA in muscle and liver, and PGC-1 α mRNA in liver. CONCLUSION The data suggest that RYGB and IT surgeries lead to enhanced GLP-1 secretion and produce similar stimulatory effects on important molecules of glucose metabolism but differential effects on key molecules of lipid oxidation in muscle and liver.
Collapse
Affiliation(s)
- Adel Pezeshki
- Department of Production Animal Health, Faculty of Veterinary Medicine, Gastrointestinal Research Group, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Prasanth K Chelikani
- Department of Production Animal Health, Faculty of Veterinary Medicine, Gastrointestinal Research Group, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
40
|
The MRC1/CD68 ratio is positively associated with adipose tissue lipogenesis and with muscle mitochondrial gene expression in humans. PLoS One 2013; 8:e70810. [PMID: 23951013 PMCID: PMC3741275 DOI: 10.1371/journal.pone.0070810] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 06/24/2013] [Indexed: 01/05/2023] Open
Abstract
Background Alternative macrophages (M2) express the cluster differentiation (CD) 206 (MCR1) at high levels. Decreased M2 in adipose tissue is known to be associated with obesity and inflammation-related metabolic disturbances. Here we aimed to investigate MCR1 relative to CD68 (total macrophages) gene expression in association with adipogenic and mitochondrial genes, which were measured in human visceral [VWAT, n = 147] and subcutaneous adipose tissue [SWAT, n = 76] and in rectus abdominis muscle (n = 23). The effects of surgery-induced weight loss were also longitudinally evaluated (n = 6). Results MCR1 and CD68 gene expression levels were similar in VWAT and SWAT. A higher proportion of CD206 relative to total CD68 was present in subjects with less body fat and lower fasting glucose concentrations. The ratio MCR1/CD68was positively associated with IRS1gene expression and with the expression of lipogenic genes such as ACACA, FASN and THRSP, even after adjusting for BMI. The ratio MCR1/CD68 in SWAT increased significantly after the surgery-induced weight loss (+44.7%; p = 0.005) in parallel to the expression of adipogenic genes. In addition, SWAT MCR1/CD68ratio was significantly associated with muscle mitochondrial gene expression (PPARGC1A, TFAM and MT-CO3). AT CD206 was confirmed by immunohistochemistry to be specific of macrophages, especially abundant in crown-like structures. Conclusion A decreased ratio MCR1/CD68 is linked to adipose tissue and muscle mitochondrial dysfunction at least at the level of expression of adipogenic and mitochondrial genes.
Collapse
|
41
|
Vijgen GHEJ, Bouvy ND, Hoeks J, Wijers S, Schrauwen P, van Marken Lichtenbelt WD. Impaired skeletal muscle mitochondrial function in morbidly obese patients is normalized one year after bariatric surgery. Surg Obes Relat Dis 2013; 9:936-41. [PMID: 23791452 DOI: 10.1016/j.soard.2013.03.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 01/08/2013] [Accepted: 03/22/2013] [Indexed: 11/17/2022]
Abstract
BACKGROUND Obesity and type 2 diabetes are associated with impaired skeletal muscle mitochondrial metabolism. As an intrinsic characteristic of an individual, skeletal muscle mitochondrial dysfunction could be a risk factor for weight gain and obesity-associated co-morbidities, such as type 2 diabetes. On the other hand, impaired skeletal muscle metabolism could be a consequence of obesity. We hypothesize that marked weight loss after bariatric surgery recovers skeletal muscle mitochondrial function. METHODS Skeletal muscle mitochondrial function as assessed by high-resolution respirometry was measured in 8 morbidly obese patients (body mass index [BMI], 41.3±4.7 kg/m(2); body fat, 48.3%±5.2%) before and 1 year after bariatric surgery (mean weight loss: 35.0±8.6 kg). The results were compared with a lean (BMI 22.8±1.1 kg/m(2); body fat, 15.6%±4.7%) and obese (BMI 33.5±4.2 kg/m(2); body fat, 34.1%±6.3%) control group. RESULTS Before surgery, adenosine diphosphate (ADP)-stimulated (state 3) respiration on glutamate/succinate was decreased compared with lean patients (9.5±2.4 versus 15.6±4.4 O2 flux/mtDNA; P<.05). One year after surgery, mitochondrial function was comparable to that of lean controls (after weight loss, 12.3±5.5; lean, 15.6±4.4 O2 flux/mtDNA). In addition, we observed an increased state 3 respiration on a lipid substrate after weight loss (10.0±3.2 versus 14.0±6.6 O2 flux/mtDNA; P< .05). CONCLUSION We conclude that impaired skeletal muscle mitochondrial function is a consequence of obesity that recovers after marked weight loss.
Collapse
Affiliation(s)
- Guy H E J Vijgen
- Department of Surgery, Maastricht University Medical Centre, Maastricht, the Netherlands; Department of Human Biology, School for Nutrition, Toxicology and Metabolism, NUTRIM, Maastricht, the Netherlands.
| | | | | | | | | | | |
Collapse
|
42
|
Gan KX, Wang C, Chen JH, Zhu CJ, Song GY. Mitofusin-2 ameliorates high-fat diet-induced insulin resistance in liver of rats. World J Gastroenterol 2013; 19:1572-1581. [PMID: 23538485 PMCID: PMC3602474 DOI: 10.3748/wjg.v19.i10.1572] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Revised: 11/13/2012] [Accepted: 01/05/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of mitofusin-2 (MFN2) on insulin sensitivity and its potential targets in the liver of rats fed with a high-fat diet (HFD).
METHODS: Rats were fed with a control or HFD for 4 or 8 wk, and were then infected with a control or an MFN2 expressing adenovirus once a week for 3 wk starting from the 9th wk. Blood glucose (BG), plasma insulin and insulin sensitivity of rats were determined at end of the 4th and 8th wk, and after treatment with different amounts of MFN2 expressing adenovirus (108, 109 or 1010 vp/kg body weight). BG levels were measured by Accu-chek Active Meter. Plasma insulin levels were analyzed by using a Rat insulin enzyme-linked immunosorbent assay kit. Insulin resistance was evaluated by measuring the glucose infusion rate (GIR) using a hyperinsulinemic euglycemic clamp technique. The expression or phosphorylation levels of MFN2 and essential molecules in the insulin signaling pathway, such as insulin receptor (INSR), insulin receptor substrate 2 (IRS2), phosphoinositide-3-kinase (PI3K), protein kinase beta (AKT2) and glucose transporter type 2 (GLUT2) was assayed by quantitative real-time polymerase chain reaction and Western-blotting.
RESULTS: After the end of 8 wk, the body weight of rats receiving the normal control diet (ND) and the HFD was not significantly different (P > 0.05). Compared with the ND group, GIR in the HFD group was significantly decreased (P < 0.01), while the levels of BG, triglycerides (TG), total cholesterol (TC) and insulin in the HFD group were significantly higher than those in the ND group (P < 0.05). Expression of MFN2 mRNA and protein in liver of rats was significantly down-regulated in the HFD group (P < 0.01) after 8 wk of HFD feeding. The expression of INSR, IRS2 and GLUT2 were down-regulated markedly (P < 0.01). Although there were no changes in PI3K-P85 and AKT2 expression, their phosphorylation levels were decreased significantly (P < 0.01). After intervention with MFN2 expressing adenovirus for 3 wk, the expression of MFN2 mRNA and protein levels were up-regulated (P < 0.01). There was no difference in body weight of rats between the groups. The levels of BG, TG, TC and insulin in rats were lower than those in the Ad group (P < 0.05), but GIR in rats infected with Ad-MFN2 was significantly increased (P < 0.01), compared with the Ad group. The expression of INSR, IRS2 and GLUT2 was increased, while phosphorylation levels of PI3K-P85 and AKT2 were increased (P < 0.01), compared with the Ad group.
CONCLUSION: HFDs induce insulin resistance, and this can be reversed by MFN2 over-expression targeting the insulin signaling pathway.
Collapse
|
43
|
Cornall LM, Mathai ML, Hryciw DH, Simcocks AC, O'Brien PE, Wentworth JM, McAinch AJ. GPR119 regulates genetic markers of fatty acid oxidation in cultured skeletal muscle myotubes. Mol Cell Endocrinol 2013; 365:108-18. [PMID: 23069642 DOI: 10.1016/j.mce.2012.10.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 10/01/2012] [Accepted: 10/03/2012] [Indexed: 12/16/2022]
Abstract
Gene knockout and agonist studies indicate that activation of the G protein-coupled receptor, GPR119, protects against diet-induced obesity and insulin resistance. It is not known if GPR119 activation in skeletal muscle mediates these effects. To address this uncertainty, we measured GPR119 expression in skeletal muscle and determined the effects of PSN632408, a GPR119 agonist, on the expression of genes and proteins required for fatty acid and glucose oxidation in cultured myotubes. GPR119 expression was readily detected in rat skeletal muscle and mRNAs were induced by 12 weeks of high-fat feeding. Treatment of cultured mouse C₂C₁₂ myotubes with 5 μM PSN632408 or 0.5 mM palmitate reduced expression of mRNAs encoding fatty acid oxidation genes to similar extents. More so, treatment with PSN632408 decreased AMPKα (Thr172 phosphorylation) activity in the absence of palmitate and ACC (Ser79 phosphorylation) activity in the presence of palmitate. In human primary myotubes PSN632408 decreased expression of PDK4 and AMPKα2 mRNA in myotubes derived from obese donors. These data suggest GPR119 activation in skeletal muscle may impair fatty acid and glucose oxidation.
Collapse
MESH Headings
- Acids, Heterocyclic/pharmacology
- Adult
- Animals
- Body Mass Index
- Cells, Cultured
- Clone Cells
- Fatty Acids, Nonesterified/metabolism
- Female
- Gene Expression Regulation/drug effects
- Genetic Markers
- Glucose/metabolism
- Humans
- Male
- Mice
- Middle Aged
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Obesity, Morbid/genetics
- Obesity, Morbid/metabolism
- Obesity, Morbid/pathology
- Oxadiazoles/pharmacology
- Random Allocation
- Rats
- Rats, Sprague-Dawley
- Receptors, G-Protein-Coupled/agonists
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
Collapse
Affiliation(s)
- L M Cornall
- Biomedical and Lifestyle Diseases Unit, School of Biomedical and Health Sciences, Victoria University, Melbourne 8001, Australia.
| | | | | | | | | | | | | |
Collapse
|
44
|
Zhang F, Kong D, Lu Y, Zheng S. Peroxisome proliferator-activated receptor-γ as a therapeutic target for hepatic fibrosis: from bench to bedside. Cell Mol Life Sci 2013; 70:259-76. [PMID: 22699820 PMCID: PMC11113701 DOI: 10.1007/s00018-012-1046-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 05/18/2012] [Accepted: 05/29/2012] [Indexed: 02/07/2023]
Abstract
Hepatic fibrosis is a dynamic chronic liver disease occurring as a consequence of wound-healing responses to various hepatic injuries. This disorder is one of primary predictors for liver-associated morbidity and mortality worldwide. To date, no pharmacological agent has been approved for hepatic fibrosis or could be recommended for routine use in clinical context. Cellular and molecular understanding of hepatic fibrosis has revealed that peroxisome proliferator-activated receptor-γ (PPARγ), the functioning receptor for antidiabetic thiazolidinediones, plays a pivotal role in the pathobiology of hepatic stellate cells (HSCs), whose activation is the central event in the pathogenesis of hepatic fibrosis. Activation of PPARγ inhibits HSC collagen production and modulates HSC adipogenic phenotype at transcriptional and epigenetic levels. These molecular insights indicate PPARγ as a promising drug target for antifibrotic chemotherapy. Intensive animal studies have demonstrated that stimulation of PPARγ regulatory system through gene therapy approaches and PPARγ ligands has therapeutic promise for hepatic fibrosis induced by a variety of etiologies. At the same time, thiazolidinedione agents have been investigated for their clinical benefits primarily in patients with nonalcoholic steatohepatitis, a common metabolic liver disorder with high potential to progress to fibrosis and liver-related death. Although some studies have shown initial promise, none has established long-term efficacy in well-controlled randomized clinical trials. This comprehensive review covers the 10-year discoveries of the molecular basis for PPARγ regulation of HSC pathophysiology and then focuses on the animal investigations and clinical trials of various therapeutic modalities targeting PPARγ for hepatic fibrosis.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 282 Hanzhong Road, Nanjing, 210029 Jiangsu China
| | - Desong Kong
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 282 Hanzhong Road, Nanjing, 210029 Jiangsu China
| | - Yin Lu
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 282 Hanzhong Road, Nanjing, 210029 Jiangsu China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210046 China
- National First-Class Key Discipline for Traditional Chinese Medicine of Nanjing University of Chinese Medicine, Nanjing, 210046 China
| | - Shizhong Zheng
- Department of Clinical Pharmacy, College of Pharmacy, Nanjing University of Chinese Medicine, 282 Hanzhong Road, Nanjing, 210029 Jiangsu China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210046 China
- National First-Class Key Discipline for Traditional Chinese Medicine of Nanjing University of Chinese Medicine, Nanjing, 210046 China
| |
Collapse
|
45
|
Prevention mechanisms of glucose intolerance and obesity by cacao liquor procyanidin extract in high-fat diet-fed C57BL/6 mice. Arch Biochem Biophys 2012; 527:95-104. [DOI: 10.1016/j.abb.2012.03.018] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 02/27/2012] [Accepted: 03/15/2012] [Indexed: 12/25/2022]
|
46
|
Bradley D, Magkos F, Klein S. Effects of bariatric surgery on glucose homeostasis and type 2 diabetes. Gastroenterology 2012; 143:897-912. [PMID: 22885332 PMCID: PMC3462491 DOI: 10.1053/j.gastro.2012.07.114] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Revised: 07/20/2012] [Accepted: 07/23/2012] [Indexed: 12/19/2022]
Abstract
Obesity is an important risk factor for type 2 diabetes mellitus (T2DM). Weight loss improves the major factors involved in the pathogenesis of T2DM, namely insulin action and beta cell function, and is considered a primary therapy for obese patients who have T2DM. Unfortunately, most patients with T2DM fail to achieve successful weight loss and adequate glycemic control from medical therapy. In contrast, bariatric surgery causes marked weight loss and complete remission of T2DM in most patients. Moreover, bariatric surgical procedures that divert nutrients away from the upper gastrointestinal tract are more successful in producing weight loss and remission of T2DM than those that simply restrict stomach capacity. Although upper gastrointestinal tract bypass procedures alter the metabolic response to meal ingestion, by increasing early postprandial plasma concentrations of glucagon-like peptide 1 and insulin, it is not clear whether these effects make an important contribution to long-term control of glycemia and T2DM once substantial surgery-induced weight loss has occurred. Nonetheless, the effects of surgery on body weight and metabolic function indicate that bariatric surgery should be part of the standard therapy for T2DM. More research is needed to advance our understanding of the physiological effects of different bariatric surgical procedures and possible weight loss-independent factors that improve metabolic function and contribute to the resolution of T2DM.
Collapse
|
47
|
Russell AP, Wada S, Vergani L, Hock MB, Lamon S, Léger B, Ushida T, Cartoni R, Wadley GD, Hespel P, Kralli A, Soraru G, Angelini C, Akimoto T. Disruption of skeletal muscle mitochondrial network genes and miRNAs in amyotrophic lateral sclerosis. Neurobiol Dis 2012; 49:107-17. [PMID: 22975021 DOI: 10.1016/j.nbd.2012.08.015] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Revised: 08/05/2012] [Accepted: 08/17/2012] [Indexed: 12/25/2022] Open
Abstract
Skeletal muscle mitochondrial dysfunction is believed to play a role in the progression and severity of amyotrophic lateral sclerosis (ALS). The regulation of transcriptional co-activators involved in mitochondrial biogenesis and function in ALS is not well known. When compared with healthy control subjects, patients with ALS, but not neurogenic disease (ND), had lower levels of skeletal muscle peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) mRNA and protein and estrogen-related receptor-α (ERRα) and mitofusin-2 (Mfn2) mRNA. PGC-1β, nuclear respiratory factor-1 (NRF-1) and Mfn1 mRNA as well as cytochrome C oxidase subunit IV (COXIV) mRNA and protein were lower in patients with ALS and ND. Both patient groups had reductions in citrate synthase and cytochrome c oxidase activity. Similar observations were made in skeletal muscle from transgenic ALS G93A transgenic mice. In vitro, PGC-1α and PGC-1β regulated Mfn1 and Mfn2 in an ERRα-dependent manner. Compared to healthy controls, miRNA 23a, 29b, 206 and 455 were increased in skeletal muscle of ALS patients. miR-23a repressed PGC-1α translation in a 3' UTR dependent manner. Transgenic mice over expressing miR-23a had a reduction in PGC-1α, cytochome-b and COXIV protein levels. These results show that skeletal muscle mitochondrial dysfunction in ALS patients is associated with a reduction in PGC-1α signalling networks involved in mitochondrial biogenesis and function, as well as increases in several miRNAs potentially implicated in skeletal muscle and neuromuscular junction regeneration. As miR-23a negatively regulates PGC-1α signalling, therapeutic inhibition of miR-23a may be a strategy to rescue PGC-1α activity and ameliorate skeletal muscle mitochondrial function in ALS.
Collapse
Affiliation(s)
- Aaron P Russell
- Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, 3125 Burwood, Australia.
| | - Shogo Wada
- Division of Regenerative Medical Engineering, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo, 113-0033 Tokyo, Japan
| | - Lodovica Vergani
- Neurosciences Department, University of Padua, 35129 Padua, Italy
| | - M Benjamin Hock
- Department of Chemical Physiology, The Scripps Research Institute,92037 La Jolla, USA
| | - Séverine Lamon
- Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, 3125 Burwood, Australia
| | - Bertrand Léger
- Clinique romande de réadaptation, 1951 Sion, Switzerland
| | - Takashi Ushida
- Division of Regenerative Medical Engineering, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo, 113-0033 Tokyo, Japan
| | - Romain Cartoni
- Clinique romande de réadaptation, 1951 Sion, Switzerland
| | - Glenn D Wadley
- Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, 3125 Burwood, Australia
| | - Peter Hespel
- Research Centre for Exercise and Health, Faculty of Kinesiology and Rehabilitation Sciences, K.U. Leuven, B-3001 Leuven, Belgium
| | - Anastasia Kralli
- Department of Chemical Physiology, The Scripps Research Institute,92037 La Jolla, USA
| | - Gianni Soraru
- Neurosciences Department, University of Padua, 35129 Padua, Italy
| | - Corrado Angelini
- Neurosciences Department, University of Padua, 35129 Padua, Italy; I.R.C.S.S. S. Camillo, 30126 Venice, Italy
| | - Takayuki Akimoto
- Division of Regenerative Medical Engineering, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo, 113-0033 Tokyo, Japan.
| |
Collapse
|
48
|
Kee AL, Isenring E, Hickman I, Vivanti A. Resting energy expenditure of morbidly obese patients using indirect calorimetry: a systematic review. Obes Rev 2012; 13:753-65. [PMID: 22568725 DOI: 10.1111/j.1467-789x.2012.01000.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The increasing proportion of acutely ill hospital patient admissions presenting with a morbidly obese body mass index (BMI ≥ 40 kg m(-2) ) as a comorbidity is an emerging clinical concern. Suboptimal food intake and malnutrition is prevalent in the acute care hospital setting. The energy requirements necessary to prevent malnutrition in acutely ill patients with morbid obesity remains unclear. The aim of this systematic review was to identify studies in the literature that have used indirect calorimetry to measure the resting energy expenditure of patients with morbid obesity to establish their minimum energy requirements and the implications for optimal feeding practices in acutely ill hospitalized patients. A total of 20 studies from PubMed, Cochrane Library and Embase met the inclusion criteria and were reviewed. All articles were graded using the Australian National Health and Medical Research Council levels of evidence and given a quality rating using the American Dietetic Association recommendations. Studies were categorized according to the mean BMI of its subjects. The most commonly measured resting energy expenditures for morbidly obese patients are between 2,000 and 3,000 kcal d(-1) (8,400-12,600 kJ d(-1) ). Activity and injury factors of acutely ill morbidly obese patients could result in significantly greater energy requirements for this patient group and are unlikely to be met by standard hospital menus. Establishing the minimum energy requirements for this population group will help inform adequate and accurate energy provision in the acute setting. Outcomes of underfeeding and overfeeding in morbidly obese patients warrant further research.
Collapse
Affiliation(s)
- A-L Kee
- School of Human Movement Studies, University of Queensland, Brisbane, Queensland, Australia
| | | | | | | |
Collapse
|
49
|
Dirksen C, Jørgensen NB, Bojsen-Møller KN, Jacobsen SH, Hansen DL, Worm D, Holst JJ, Madsbad S. Mechanisms of improved glycaemic control after Roux-en-Y gastric bypass. Diabetologia 2012; 55:1890-901. [PMID: 22538359 DOI: 10.1007/s00125-012-2556-7] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 02/21/2012] [Indexed: 12/13/2022]
Abstract
Roux-en-Y gastric bypass (RYGB) greatly improves glycaemic control in morbidly obese patients with type 2 diabetes, in many even before significant weight loss. Understanding the responsible mechanisms may contribute to our knowledge of the pathophysiology of type 2 diabetes and help identify new drug targets or improve surgical techniques. This review summarises the present knowledge based on pathophysiological studies published during the last decade. Taken together, two main mechanisms seem to be responsible for the early improvement in glycaemic control after RYGB: (1) an increase in hepatic insulin sensitivity induced, at least in part, by energy restriction and (2) improved beta cell function associated with an exaggerated postprandial glucagon-like peptide 1 secretion owing to the altered transit of nutrients. Later a weight loss induced improvement in peripheral insulin sensitivity follows.
Collapse
Affiliation(s)
- C Dirksen
- Department of Endocrinology 541, Hvidovre Hospital, University of Copenhagen, Kettegaard Allé 30, 2650 Hvidovre, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Tinahones FJ, Moreno-Santos I, Vendrell J, Chacon MR, Garrido-Sanchez L, García-Fuentes E, Macias-González M. The retinoic acid receptor-related orphan nuclear receptor γ1 (RORγ1): a novel player determinant of insulin sensitivity in morbid obesity. Obesity (Silver Spring) 2012; 20:488-97. [PMID: 21904299 DOI: 10.1038/oby.2011.267] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The orphan nuclear receptors (ONRs), retinoic acid receptor-related orphan receptor γ-1 (RORγ1) and peroxisome proliferator-activated receptor γ-2 (PPARγ2), are central mediators controlling adipocyte (AD) differentiation. Through their distinct tissue distribution and specific target gene activation, ONRs control diverse aspects of fatty acid metabolism and insulin sensitivity. Adding further complexity, obesity begets resistance to insulin signals and can ultimately result in diabetes. In this study, we investigate whether there are differences in the RORγ1 and PPARγ2 expression in visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT) from morbid obesity (MO) individuals either insulin resistant (high-IR MO) or insulin sensitivity (low-IR MO). Our results indicate for the first time in human the RORγ1 mRNA and protein expression levels and activation with coactivator, such as peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) were higher in the VAT from high-IR MO. In contrast, PPARγ2 expression and activation were higher in the VAT from low-IR MO. In this way, we have also found a positive association between RORγ1 mRNA and protein expression with many components of metabolic syndrome, with a strong dependence of insulin and HOMA(IR) index in VAT, but not in SAT. Our data suggest that RORγ1 may be added to the growing list of nuclear receptors in adipose tissue use to modulate the insulin resistance associated to the obesity. Measurement of RORγ1 and PPARγ2 in adipose tissue might be useful for evaluating the outcomes of various clinical interventions for obesity-related diabetes type II.
Collapse
Affiliation(s)
- Francisco J Tinahones
- Laboratorio de Investigación Biomédica, Servicio de Endocrinologia Nutricion, Hospital Virgen de la Victoria (Fundacion IMABIS), Malaga, Spain
| | | | | | | | | | | | | |
Collapse
|